Language selection

Search

Patent 2716906 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2716906
(54) English Title: MICRORNA-BASED METHODS AND COMPOSITIONS FOR THE DIAGNOSIS, PROGNOSIS AND TREATMENT OF GASTRIC CANCER
(54) French Title: PROCEDES ET COMPOSITIONS FONDES SUR MICROARN POUR LE DIAGNOSTIC, LE PRONOSTIC ET LE TRAITEMENT DU CANCER DE L'ESTOMAC
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07H 21/02 (2006.01)
  • C12N 15/113 (2010.01)
  • A61K 31/7105 (2006.01)
  • A61P 1/04 (2006.01)
  • A61P 35/00 (2006.01)
  • C40B 40/06 (2006.01)
  • G01N 33/53 (2006.01)
  • C40B 30/04 (2006.01)
  • C12Q 1/68 (2006.01)
(72) Inventors :
  • CROCE, CARLO M. (United States of America)
  • PETROCCA, FABIO (United States of America)
  • VECCHIONE, ANDREA (United States of America)
(73) Owners :
  • THE OHIO STATE UNIVERSITY RESEARCH FOUNDATION (United States of America)
(71) Applicants :
  • THE OHIO STATE UNIVERSITY RESEARCH FOUNDATION (United States of America)
(74) Agent: RIDOUT & MAYBEE LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2009-02-27
(87) Open to Public Inspection: 2009-09-03
Examination requested: 2014-02-19
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2009/035458
(87) International Publication Number: WO2009/108853
(85) National Entry: 2010-08-26

(30) Application Priority Data:
Application No. Country/Territory Date
61/067,445 United States of America 2008-02-28

Abstracts

English Abstract



Methods and compositions for the diagnosis, prognosis and/or treatment of
gastric cancer associated diseases are
disclosed.


French Abstract

L'invention concerne des procédés et des compositions pour le diagnostic, le pronostic et/ou le traitement de maladies associées au cancer de l'estomac.

Claims

Note: Claims are shown in the official language in which they were submitted.



CLAIMS
What is claimed is:

1. A method of diagnosing whether a subject has, or is at risk for developing
a
gastric-related disorder, determining a prognosis of a subject with gastric
cancer and/or
related disorder, and/or treating the subject who has such disorder,
comprising
measuring the level of at least one biomarker in a test sample from the
subject,
wherein an alteration in the level of the biomarker in the test sample,
relative to the level of a corresponding biomarker in a control sample, is
indicative of the
subject either having, or being at risk for developing, the disorder.

2. The method of Claim 1, wherein the level of the at least one biomarker in
the test sample is less than the level of the corresponding biomarker in the
control sample.
3. The method of Claim 1, wherein the level of the at least one biomarker in
the test sample is greater than the level of the corresponding biomarker in
the control
sample.

4. The method of Claim 1, wherein the at least one biomarker differentially
expressed is selected from the group listed in Figure 13 - Table 1.

5. The method of Claim 4, wherein the disorder comprises chronic gastritis
and at least one biomarker is selected from the group consisting of: miR-1 and
miR-155
that are up-regulated.

6. The method of Claim 4, wherein the disorder comprises chronic gastritis
and at least one biomarker is selected from the group consisting of: miR-205,
miR-203,
miR-202, miR-20 and miR-26b that are down-regulated.

7. The method of Claim 1, wherein the at least one biomarker differentially
expressed is selected from the group listed in Figure 14 - Table 2.

69


8. The method of Claim 7, wherein the disorder comprises gastric
adenocarcinoma and at least one biomarker is selected from the group
consisting of miR-
21, miR-223, miR-25, miR-17-5-p, miR-125b, miR-181b, miR-106a, miR-107, miR-
92,
miR-103, miR-221, miR-93, miR-100, miR-181, miR-106b, miR-191, miR-214, miR-
130,
miR-342, miR-222, miR-320 and miR-99b that are up-regulated.

9. The method of Claim 7, wherein the disorder comprises gastric
adenocarcinoma and at least one biomarker is selected from the group
consisting of: miR-
136, miR-218, miR-212, miR-96, miR-339 and miR-130b that are down-regulated.

10. The method of Claim 1, wherein the at least one biomarker differentially
expressed is selected from the group listed in Figure 16 - Table 3: miR-21,
miR-223,
miR-25, miR-92, miR-107, miR-93, miR-106b, miR-17-5p, miR-181b and miR-106a.

11. The method of Claim 1, wherein the at least one biomarker comprises the
miR-160b-25 cluster: miR-106b, miR-93 and miR-25.

12. Use of at least one biomarker comprising the miR-160b-25 cluster: miR-
106b, miR-93 and miR-25, in the modulation of expression of one or more of the
genes
listed in Figure 18 - Table 6: PHLPPL, GM632, ALX4, PLEKHM1, JOSD1, ZFPM2,
GATAD2B, ZNF238, ATXN1, NEUROD1, BCL2L11, KLF12, UBE2W, OSBPL5,
SNF1LK, PCAF, PAPOLA, and CFL2.

13. A method for regulating E2F1 expression in a subject in need thereof,
comprising administering an effective amount of miR-106b and/or miR-93, or a
functional
variant thereof, sufficient to modulate expression of E2F1.

14. Use of miR-106b and miR-93 to regulate E2F1 expression in a subject in
need thereof.

15. A method modulating a TGFE tumor suppressor pathway that interferes
with expression of CDKN1A (p21Waf1/Cip1 ) and/or BCL2L11(Bim), comprising up-
regulating one or more of miR-106b, miR-93 and miR-25.



16. Use of miR-106b-25 cluster in E2F1 post-transcriptional regulation and
modulation of development of TGFE resistance in gastric cancer.

17. A method for controlling E2F1 expression in a subject in need thereof,
comprising modulating levels of miR-106b and miR-93 in the subject.

18. Use of E2F1 to regulates miR-106b-25 expression in parallel with Mcm7,
in a subject in need thereof.

19. A method for controlling the rate of E2F1 protein synthesis, preventing
its
excessive accumulation in a subject in need thereof, comprising modulating
levels of the
miR-106b-25 cluster in the subject.

20. Use of miR-106b and miR-93 to impair TGFE-induced cell cycle arrest in a
subject in need thereof.

21. Use of miR-106b and miR-93 to interfere with TGFO-induced cell cycle
arrest by inhibiting expression of p21 at a post-transcriptional level in a
subject in need
thereof.

22. Use of miR-25 in cooperation with miR-106b and miR-93 in preventing the
onset of TGFO-induced apoptosis, in a subject in need thereof.

23. Method for modulating expression of the miR-106b-25 cluster to prevent
protection of gastric cancer cells from apoptosis in a subject in need
thereof.

24. A distinct microRNA expression signature in gastric cancer comprising
alterations in the expression of one or more biomarkers that regulate tumor
microRNA
processing.

25. A method for influencing transcript abundance and/or protein expression of
target mRNAs in gastric cancer, comprising deregulating one or more microRNAs
in a

71


subject in need thereof.

26. The method of the preceding Claim, comprising inhibiting the protein
expression of cancer-related genes.

27. The method of the preceding claim, comprising altering expression of one
or more of miR-106b, miR-93 and miR-25 to inhibit the protein expression of
cancer-
related genes.

28. Use of a large-scale gene expression profiling of both microRNAs and
protein-encoding RNAs to identify alterations in microRNA function that occur
in human
gastric cancer.

29. The method of Claim 1, comprising determining the prognosis of a subject
with gastric cancer, comprising measuring the level of at least one biomarker
in a test
sample from the subject, wherein:
i) the biomarker is associated with an adverse prognosis in such cancer; and
ii) an alteration in the level of the at least one biomarker in the test
sample, relative
to the level of a corresponding biomarker in a control sample, is indicative
of an adverse
prognosis.

30. The method of Claim 1, comprising diagnosing whether a subject has, or is
at risk for developing, gastric cancer, comprising:
1) reverse transcribing RNA from a test sample obtained from the subject to
provide a set of target oligodeoxynucleotides;
2) hybridizing the target oligodeoxynucleotides to a microarray comprising
miRNA-specific probe oligonucleotides to provide a hybridization profile for
the test
sample; and
3) comparing the test sample hybridization profile to a hybridization profile
generated from a control sample, wherein an alteration in the signal of at
least one miRNA
is indicative of the subject either having, or being at risk for developing,
such cancer.

31. The method of the preceding Claim, wherein the signal of at least one
72


miRNA, relative to the signal generated from the control sample, is down-
regulated,
and/or wherein the signal of at least one miRNA, relative to the signal
generated from the
control sample, is up-regulated.

32. The method of the preceding Claim, wherein an alteration in the signal of
at
least one biomarker selected from the group listed in: Table 13, Table 14 and
Table 16,
are indicative of the subject either having, or being at risk for developing,
such cancer with
an adverse prognosis.

33. A biomarker of a gastric disorder or disease, comprising one or more of:
miR-106b, miR-93 and mir-25.

34. A method for regulating protein expression in gastric cancer cells,
comprising modulating the expression of one or more of: miR-106b, miR-93 and
mir-25 in
the gastric cancer cells.

35. A composition for modulating expression of one or more of E2F1,
CDKN1A(p21Waf1Cip1) and BCL2L11(Bim) in gastric cancer cells, the composition
comprising one or more of: miR-106b, miR-93 and mir-25, or functional variants
thereof.

36. A method for regulating one or more of E2F1 and p21/WAF1 protein levels
in a subject in need thereof, comprising using one or more of: miR-106b, miR-
93 and mir-
25, or functional variants thereof.

37. A composition comprising antisense miR-106b useful to increase
p21/WAF1 and/or E2F1 protein levels in gastric cancer cells in a subject in
need thereof.
38. A method of treating gastric cancer in a subject who has a gastric cancer
in
which at least one biomarker is down-regulated or up-regulated in the cancer
cells of the
subject relative to control cells, comprising:
1) when the at least one biomarker is down-regulated in the cancer cells,
administering to the subject an effective amount of at least one isolated
biomarker, or an
isolated variant or biologically-active fragment thereof, such that
proliferation of cancer

73


cells in the subject is inhibited; or
2) when the at least one biomarker is up-regulated in the cancer cells,
administering to the subject an effective amount of at least one compound for
inhibiting
expression of the at least one biomarker, such that proliferation of cancer
cells in the
subject is inhibited.

39. A method of treating gastric cancer in a subject, comprising:
1) determining the amount of at least one biomarker in gastric cancer cells,
relative
to control cells; and
2) altering the amount of biomarker expressed in the gastric cancer cells by:
i) administering to the subject an effective amount of at least one isolated
biomarker, if the amount of the biomarker expressed in the cancer cells is
less than the
amount of the biomarker expressed in control cells; or
ii) administering to the subject an effective amount of at least one
compound for inhibiting expression of the at least one biomarker, if the
amount of the
biomarker expressed in the cancer cells is greater than the amount of the
biomarker
expressed in control cells.

40. A pharmaceutical composition for treating gastric cancer, comprising at
least one isolated biomarker, and a pharmaceutically-acceptable carrier.

41. The pharmaceutical composition of the preceding Claim, wherein the at
least one isolated biomarker corresponds to a biomarker that is down-regulated
in gastric
cancer cells relative to control cells.

42. The pharmaceutical composition of the preceding Claim; comprising at
least one miR expression-inhibitor compound and a pharmaceutically-acceptable
carrier.
43. A method of identifying an anti-gastric cancer agent, comprising providing
a test agent to a cell and measuring the level of at least one biomarker
associated with
decreased expression levels in gastric cancer cells, wherein an increase in
the level of the
biomarker in the cell, relative to a control cell, is indicative of the test
agent being an anti-
gastric cancer agent.

74



44. A method of identifying an anti-gastric cancer agent, comprising providing

a test agent to a cell and measuring the level of at least one biomarker
associated with
increased expression levels in gastric cancer cells, wherein a decrease in the
level of the
biomarker in the cell, relative to a control cell, is indicative of the test
agent being an anti-
cancer agent.

45. A method of assessing the effectiveness of a therapy to prevent, diagnose
and/or
treat a gastric cancer associated disease, comprising:
i) subjecting an animal to a therapy whose effectiveness is being assessed,
and
ii) determining the level of effectiveness of the treatment being tested in
treating or
preventing the disease, by evaluating at least one biomarker listed in one or
more of Tables 13,
14 and 16.

46. The method of the preceding Claim, wherein the candidate therapeutic
agent comprises one or more of: pharmaceutical compositions, nutraceutical
compositions,
and homeopathic compositions.

47. The method of the preceding Claim, wherein the therapy being assessed is
for
use in a human subject.

48. An article of manufacture comprising: at least one capture reagent that
binds to a marker for a gastric cancer associated disease comprising at least
one biomarker
listed in one or more of Tables 13, 14 and 16.

49. A kit for screening for a candidate compound for a therapeutic agent to
treat a gastric cancer associated disease, wherein the kit comprises: one or
more reagents
of at least one biomarker listed in one or more of Tables 13, 14 and 16, and a
cell expressing
at least one biomarker.

50. The kit of the preceding Claim, wherein the presence of the biomarker is
detected using a reagent comprising an antibody or an antibody fragment which
specifically binds with at least one biomarker.



51. Use of an agent that interferes with a gastric cancer associated disease
response signaling pathway, for the manufacture of a medicament for treating,
preventing,
reversing or limiting the severity of the disease complication in an
individual, wherein the
agent comprises at least one biomarker listed in one or more of Tables 13, 14
and 16.

52. A method of treating, preventing, reversing or limiting the severity of a
gastric cancer associated disease complication in an individual in need
thereof,
comprising: administering to the individual an agent that interferes with at
least a gastric
cancer associated disease response cascade, wherein the agent comprises at
least one
biomarker listed in one or more of Tables 13, 14 and 16.

53. Use of an agent that interferes with at least a gastric cancer associated
disease response cascade, for the manufacture of a medicament for treating,
preventing,
reversing or limiting the severity of a gastric cancer-related disease
complication in an
individual, wherein the agent comprises at least one biomarker listed in one
or more of
Tables 13, 14 and 16.

54. A composition comprising an antisense inhibitor of one or more of miR-
1o6b, miR-93 and miR-25.

55. A method of treating a gastric disorder in a subject in need thereof,
comprising administering to a subject a therapeutically effective amount of
the
composition of the preceding Claim.

56. The method of the preceding Claim, wherein the composition is
administered prophylactically.

57. The method of the preceding Claim, wherein administration of the
composition delays the onset of one or more symptoms of gastric cancer.

58. The method of the preceding Claim, wherein administration of the
composition inhibits development of gastric cancer.

76



59. The method of the preceding Claim, wherein administration of the
composition inhibits tumor growth.

60. A method for detecting the presence of gastric cancer in a biological
sample, comprising:
i) exposing the biological sample suspected of containing gastric cancer to a
marker therefor; and
ii) detecting the presence or absence of the marker, if any, in the sample.
61. The method of the preceding Claim, wherein the marker includes a
detectable label.

62. The method of the preceding Claim, further comprising comparing the
amount of the marker in the biological sample from the subject to an amount of
the marker
in a corresponding biological sample from a normal subject.

63. The method of the preceding Claim, further comprising collecting a
plurality of biological samples from a subject at different time points and
comparing the
amount of the marker in each biological sample to determine if the amount of
the marker
is increasing or decreasing in the subject over time.

64. A method for treating a gastric cancer in a subject, the method
comprising:
gastric cancer receptor agonist.

65. The method of the preceding Claim, wherein the receptor agonist is an
antisense inhibitor of one or more of: miR-106b, miR-93 and miR-25.

66. A use, to manufacture a drug for the treatment of gastric cancer,
comprised
of a nucleic acid molecule chosen from among the miR shown in Tables 13, 14
and 16, a
sequence derived therefrom, a complementary sequence from such miR and a
sequence
derived from such a complementary sequence.


77


67. The use according to the preceding Claim, wherein the drug comprises a
nucleic acid molecule presenting a sequence chosen from among: miRs listed in
Tables
13, 14 and 16, a sequence derived from such miRs, the complementary sequence
of such
miRs, and a sequence derived from such a complementary sequence.

68. An in vitro method to identify effective therapeutic agents or
combinations
of therapeutic agents to induce the differentiation of gastric cancer cells,
the method
comprising the stages of:

i) culturing of cells derived from a gastric tumor,
ii) adding at least one compound to the culture medium of the cell line,
iii) analyzing the evolution of the level of expression of at least one miR
between
stages (i) and (ii), and
iv) identifying compounds or combinations of compounds inducing a change in
the
level of expression of the miR between stages (i) and (ii).

69. The method of the preceding Claim, wherein stage (iii) includes the
analysis of the level of expression of at least one miR.

70. The method of the preceding Claim, wherein stage (iv) includes the
identification of the compounds or combinations of compounds modulating the
level of
expression of at least one miR.

71. The method of the preceding Claim, wherein stage (iv) includes the
identification of compounds or combinations of compounds reducing the level of
expression of at least one miR.

72. The method of the preceding Claim, wherein the compound is a therapeutic
agent for the treatment of cancer.

78

Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
TITLE
MicroRNA-Based Methods and Compositions for the
Diagnosis, Prognosis and Treatment of Gastric Cancer
Inventors: Carlo M. Croce, Fabio Petrocca, Andrea Vecchione
CROSS-REFERENCE TO RELATED APPLICATIONS
[00001] This application claims the benefit of United States Provisional
Application
Number 61/067,445, filed February 28, 2008, the entire disclosure of which is
expressly
incorporated herein by reference.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
[00002] This invention was made with government support under the NCI Grant
Number(s)
CA76259 and CA8134. The government has certain rights in this invention.

TECHNICAL FIELD AND
INDUSTRIAL APPLICABILITY OF THE INVENTION
[00003] This invention relates generally to the field of molecular biology.
Certain aspects
of the invention include application in diagnostics, therapeutics, and
prognostics of gastric
cancer related disorders.

BACKGROUND OF THE INVENTION
[00004] There is no admission that the background art disclosed in this
section legally
constitutes prior art.
[00005] Although the incidence of gastric cancer declined in Western countries
from the
1940s to the 1980s, it remains a major public health problem throughout the
world, being the
second most widely diagnosed malignancy worldwide and cause of 12% of all
cancer-related
deaths each year (Uemura et al., 2001). Over 95% of gastric tumors are
adenocarcinomas
histologically classified either as intestinal or diffuse type (Lauren P,
1965). The evolution of
intestinal tumors has been characterized as progressing through a number of
sequential steps.
Among the others, two events are characteristic of gastric tumorigenesis:
upregulation of
E2F1 (Suzuki et al., 1999) and development of TGFE resistance (Ju et al.,
2003; Park et al.,
1994).

1


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[00006] E2F1 is a master regulator of cell cycle that promotes the G1/S
transition
transactivating a variety of genes involved in chromosomal DNA replication,
including its
own promoter (DeGregori, 2002). While overexpression of E2F1 is an oncogenic
event per se
that predisposes cells to transformation (Pierce et al., 1999) it also
represents a potent
apoptotic signal when occurring over a critical threshold (Lazzerini Denchi et
al., 2005).
[00007] On the other hand, Transforming Growth Factor-beta (TGF(3 - is a
cytokine playing
a major role within the so-called morphogenetic program, a complex system of
crosstalk
between the epithelial and the stromal compartments that guides
gastrointestinal cells towards
proliferation, differentiation or apoptosis (van den Brink and Offerhaus,
2007).
[00008] In spite of considerable research into therapies to treat these
diseases, they remain
difficult to diagnose and treat effectively, and the mortality observed in
patients indicates that
improvements are needed in the diagnosis, treatment and prevention of the
disease.

SUMMARY OF THE INVENTION
[00009] In a first aspect, there is provided herein a method of diagnosing
whether a subject
has, or is at risk for developing a gastric-related disorder, determining a
prognosis of a subject
with gastric cancer and/or related disorder, and/or treating the subject who
has such disorder,
comprising: measuring the level of at least one biomarker in a test sample
from the subject,
wherein an alteration in the level of the biomarker in the test sample,
relative to the level of a
corresponding biomarker in a control sample, is indicative of the subject
either having, or
being at risk for developing, the disorder.
[00010] In certain embodiments, the level of the at least one biomarker in the
test sample is
less than the level of the corresponding biomarker in the control sample.
[00011] In certain embodiments, the level of the at least one biomarker in the
test sample is
greater than the level of the corresponding biomarker in the control sample.
[00012] In certain embodiments, the at least one biomarker differentially
expressed is
selected from the group listed in Figure 13 - Table 1.
[00013] In certain embodiments, the disorder comprises chronic gastritis and
at least one
biomarker is selected from the group consisting of: miR-1 and miR-155 that are
up-regulated.
[00014] In certain embodiments, the disorder comprises chronic gastritis and
at least one
biomarker is selected from the group consisting of: miR-205, miR-203, miR-202,
miR-20 and
miR-26b that are down-regulated.
[00015] In certain embodiments, the at least one biomarker differentially
expressed is
2


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
selected from the group listed in Figure 14 - Table 2.
[00016] In certain embodiments, the disorder comprises gastric adenocarcinoma
and at
least one biomarker is selected from the group consisting of miR-21, miR-223,
miR-25, miR-
17-5-p, miR-125b, miR-181b, miR-106a, miR-107, miR-92, miR-103, miR-221, miR-
93,
miR-100, miR-181, miR-106b, miR-191, miR-214, miR-130, miR-342, miR-222, miR-
320
and miR-99b that are up-regulated.
[00017] In certain embodiments, the disorder comprises gastric adenocarcinoma
and at
least one biomarker is selected from the group consisting of: miR-136, miR-
218, miR-212,
miR-96, miR-339 and miR-130b that are down-regulated.
[00018] In certain embodiments, the at least one biomarker differentially
expressed is
selected from the group listed in Figure 16 - Table 3: miR-21, miR-223, miR-
25, miR-92,
miR-107, miR-93, miR-106b, miR-17-5p, miR-181b and miR-106a.
[00019] In certain embodiments, the at least one biomarker comprises the miR-
160b-25
cluster: miR-106b, miR-93 and miR-25.
[00020] In another aspect, there is provided herein use of at least one
biomarker comprising
the miR-160b-25 cluster: miR-106b, miR-93 and miR-25, in the modulation of
expression of
one or more of the genes listed in Figure 18 - Table 6: PHLPPL, GM632, ALX4,
PLEKHMI,
JOSD1, ZFPM2, GATAD2B, ZNF238, ATXN1, NEUROD1, BCL2L11, KLF12, UBE2W,
OSBPL5, SNFILK, PCAF, PAPOLA, and CFL2.
[00021] In another aspect, there is provided herein a method for regulating
E2F1
expression in a subject in need thereof, comprising administering an effective
amount of miR-
106b and/or miR-93, or a functional variant thereof, sufficient to modulate
expression of
E2F1.
[00022] In another aspect, there is provided herein use of miR-106b and miR-93
to regulate
E2F1 expression in a subject in need thereof.
[00023] In another aspect, there is provided herein a method modulating a TGFE
tumor
suppressor pathway that interferes with expression of CDKNIA (p21Wafl/Cipl )
and/or
BCL2L11 (Bim), comprising up-regulating one or more of miR-106b, miR-93 and
miR-25.
[00024] In another aspect, there is provided herein use of miR-106b-25 cluster
in E2F1
post-transcriptional regulation and modulation of development of TGFE
resistance in gastric
cancer.
[00025] In another aspect, there is provided herein a method for controlling
E2F1
expression in a subject in need thereof, comprising modulating levels of miR-
106b and miR-
3


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
93 in the subject.
[00026] In another aspect, there is provided herein use of E2F1 to regulates
miR-106b-25
expression in parallel with Mcm7, in a subject in need thereof.
[00027] In another aspect, there is provided herein a method for controlling
the rate of
E2F1 protein synthesis, preventing its excessive accumulation in a subject in
need thereof,
comprising modulating levels of the miR-106b-25 cluster in the subject.
[00028] In another aspect, there is provided herein use of miR-106b and miR-93
to impair
TGFE-induced cell cycle arrest in a subject in need thereof.
[00029] In another aspect, there is provided herein use of miR-106b and miR-93
to
interfere with TGFO-induced cell cycle arrest by inhibiting expression of p21
at a post-
transcriptional level in a subject in need thereof.
[00030] In another aspect, there is provided herein use of miR-25 in
cooperation with miR-
106b and miR-93 in preventing the onset of TGFO-induced apoptosis, in a
subject in need
thereof.
[00031] In another aspect, there is provided herein a method for modulating
expression of
the miR-106b-25 cluster to prevent protection of gastric cancer cells from
apoptosis in a
subject in need thereof.
[00032] In another aspect, there is provided herein a distinct microRNA
expression
signature in gastric cancer comprising alterations in the expression of one or
more biomarkers
that regulate tumor microRNA processing.
[00033] In another aspect, there is provided herein a method for influencing
transcript
abundance and/or protein expression of target mRNAs in gastric cancer,
comprising
deregulating one or more microRNAs in a subject in need thereof.
[00034] In certain embodiments, the method further comprises inhibiting the
protein
expression of cancer-related genes.
[00035] In certain embodiments, the method further comprises altering
expression of one or
more of miR- 106b, miR-93 and miR-25 to inhibit the protein expression of
cancer-related
genes.
[00036] In another aspect, there is provided herein use of a large-scale gene
expression
profiling of both microRNAs and protein-encoding RNAs to identify alterations
in microRNA
function that occur in human gastric cancer.
[00037] The method of Claim 1, comprising determining the prognosis of a
subject with
gastric cancer, comprising measuring the level of at least one biomarker in a
test sample from
4


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
the subject, wherein: i) the biomarker is associated with an adverse prognosis
in such cancer;
and ii) an alteration in the level of the at least one biomarker in the test
sample, relative to the
level of a corresponding biomarker in a control sample, is indicative of an
adverse prognosis.
[00038] In certain embodiments, the method further comprises diagnosing
whether a
subject has, or is at risk for developing, gastric cancer, comprising: 1)
reverse transcribing
RNA from a test sample obtained from the subject to provide a set of target
oligodeoxynucleotides; 2) hybridizing the target oligodeoxynucleotides to a
microarray
comprising miRNA-specific probe oligonucleotides to provide a hybridization
profile for the
test sample; and 3) comparing the test sample hybridization profile to a
hybridization profile
generated from a control sample, wherein an alteration in the signal of at
least one miRNA is
indicative of the subject either having, or being at risk for developing, such
cancer.
[00039] In certain embodiments, the signal of at least one miRNA, relative to
the signal
generated from the control sample, is down-regulated, and/or wherein the
signal of at least one
miRNA, relative to the signal generated from the control sample, is up-
regulated.
[00040] In certain embodiments, an alteration in the signal of at least one
biomarker
selected from the group listed in: Table 13, Table 14 and Table 16, are
indicative of the
subject either having, or being at risk for developing, such cancer with an
adverse prognosis.
[00041] In another aspect, there is provided herein a biomarker of a gastric
disorder or
disease, comprising one or more of: miR-106b, miR-93 and mir-25.
[00042] In another aspect, there is provided herein a method for regulating
protein
expression in gastric cancer cells, comprising modulating the expression of
one or more of:
miR-106b, miR-93 and mir-25 in the gastric cancer cells.
[00043] In another aspect, there is provided herein a composition for
modulating
expression of one or more of E2F1, CDKNIA (p2lWaf1Cipl) and BCL2L11 (Bim) in
gastric
cancer cells, the composition comprising one or more of: miR-106b, miR-93 and
mir-25, or
functional variants thereof.
[00044] In another aspect, there is provided herein a method for regulating
one or more of
E2F1 and p21/WAF1 protein levels in a subject in need thereof, comprising
using one or more
of: miR-106b, miR-93 and mir-25, or functional variants thereof.
[00045] In another aspect, there is provided herein a composition comprising
antisense
miR-106b useful to increase p21/WAF1 and/or E2F1 protein levels in gastric
cancer cells in a
subject in need thereof.
[00046] In another aspect, there is provided herein a method of treating
gastric cancer in a


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
subject who has a gastric cancer in which at least one biomarker is down-
regulated or up-
regulated in the cancer cells of the subject relative to control cells,
comprising: 1) when the at
least one biomarker is down-regulated in the cancer cells, administering to
the subject an
effective amount of at least one isolated biomarker, or an isolated variant or
biologically-
active fragment thereof, such that proliferation of cancer cells in the
subject is inhibited; or 2)
when the at least one biomarker is up-regulated in the cancer cells,
administering to the
subject an effective amount of at least one compound for inhibiting expression
of the at least
one biomarker, such that proliferation of cancer cells in the subject is
inhibited.
[00047] In another aspect, there is provided herein a method of treating
gastric cancer in a
subject, comprising: 1) determining the amount of at least one biomarker in
gastric cancer
cells, relative to control cells; and 2) altering the amount of biomarker
expressed in the gastric
cancer cells by: i) administering to the subject an effective amount of at
least one isolated
biomarker, if the amount of the biomarker expressed in the cancer cells is
less than the amount
of the biomarker expressed in control cells; or ii) administering to the
subject an effective
amount of at least one compound for inhibiting expression of the at least one
biomarker, if the
amount of the biomarker expressed in the cancer cells is greater than the
amount of the
biomarker expressed in control cells.
[00048] In another aspect, there is provided herein a pharmaceutical
composition for
treating gastric cancer, comprising at least one isolated biomarker, and a
pharmaceutically-
acceptable carrier.
[00049] In certain embodiments, the at least one isolated biomarker
corresponds to a
biomarker that is down-regulated in gastric cancer cells relative to control
cells.
[00050] In certain embodiments, the pharmaceutical composition comprises at
least one
miR expression-inhibitor compound and a pharmaceutically-acceptable carrier.
[00051] In another aspect, there is provided herein a method of identifying an
anti-gastric
cancer agent, comprising providing a test agent to a cell and measuring the
level of at least
one biomarker associated with decreased expression levels in gastric cancer
cells, wherein an
increase in the level of the biomarker in the cell, relative to a control
cell, is indicative of the
test agent being an anti-gastric cancer agent.
[00052] In another aspect, there is provided herein a method of identifying an
anti-gastric
cancer agent, comprising providing a test agent to a cell and measuring the
level of at least
one biomarker associated with increased expression levels in gastric cancer
cells, wherein a
decrease in the level of the biomarker in the cell, relative to a control
cell, is indicative of the
6


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
test agent being an anti- cancer agent.
[00053] In another aspect, there is provided herein a method of assessing the
effectiveness
of a therapy to prevent, diagnose and/or treat a gastric cancer associated
disease, comprising:
i) subjecting an animal to a therapy whose effectiveness is being assessed,
and ii) determining
the level of effectiveness of the treatment being tested in treating or
preventing the disease, by
evaluating at least one biomarker listed in one or more of Tables 13, 14 and
16.
[00054] The method of the preceding Claim, wherein the candidate therapeutic
agent
comprises one or more of: pharmaceutical compositions, nutraceutical
compositions, and
homeopathic compositions.
[00055] In certain embodiments, the therapy being assessed is for use in a
human subject.
[00056] In another aspect, there is provided herein an article of manufacture
comprising: at
least one capture reagent that binds to a marker for a gastric cancer
associated disease
comprising at least one biomarker listed in one or more of Tables 13, 14 and
16.
[00057] In another aspect, there is provided herein a kit for screening for a
candidate
compound for a therapeutic agent to treat a gastric cancer associated disease,
wherein the kit
comprises: one or more reagents of at least one biomarker listed in one or
more of Tables 13,
14 and 16, and a cell expressing at least one biomarker.
[00058] In certain embodiments, the presence of the biomarker is detected
using a reagent
comprising an antibody or an antibody fragment which specifically binds with
at least one
biomarker.
[00059] In another aspect, there is provided herein use of an agent that
interferes with a
gastric cancer associated disease response signaling pathway, for the
manufacture of a
medicament for treating, preventing, reversing or limiting the severity of the
disease
complication in an individual, wherein the agent comprises at least one
biomarker listed in one
or more of Tables 13, 14 and 16.
[00060] In another aspect, there is provided herein a method of treating,
preventing,
reversing or limiting the severity of a gastric cancer associated disease
complication in an
individual in need thereof, comprising: administering to the individual an
agent that interferes
with at least a gastric cancer associated disease response cascade, wherein
the agent comprises
at least one biomarker listed in one or more of Tables 13, 14 and 16.
[00061] In another aspect, there is provided herein use of an agent that
interferes with at
least a gastric cancer associated disease response cascade, for the
manufacture of a
medicament for treating, preventing, reversing or limiting the severity of a
gastric cancer-

7


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
related disease complication in an individual, wherein the agent comprises at
least one
biomarker listed in one or more of Tables 13, 14 and 16.
[00062] In another aspect, there is provided herein a composition comprising
an antisense
inhibitor of one or more of miR-lo6b, miR-93 and miR-25.
[00063] In another aspect, there is provided herein a method of treating a
gastric disorder in
a subject in need thereof, comprising administering to a subject a
therapeutically effective
amount of the composition.
[00064] In certain embodiments, the composition is administered
prophylactically.
[00065] In certain embodiments, administration of the composition delays the
onset of one
or more symptoms of gastric cancer.
[00066] In certain embodiments, administration of the composition inhibits
development of
gastric cancer.
[00067] In certain embodiments, administration of the composition inhibits
tumor growth.
[00068] In another aspect, there is provided herein a method for detecting the
presence of
gastric cancer in a biological sample, comprising: i) exposing the biological
sample suspected
of containing gastric cancer to a marker therefor; and ii) detecting the
presence or absence of
the marker, if any, in the sample.
[00069] In certain embodiments, the marker includes a detectable label.
[00070] In certain embodiments, the method further comprises comparing the
amount of
the marker in the biological sample from the subject to an amount of the
marker in a
corresponding biological sample from a normal subject.
[00071] In certain embodiments, the method further comprises collecting a
plurality of
biological samples from a subject at different time points and comparing the
amount of the
marker in each biological sample to determine if the amount of the marker is
increasing or
decreasing in the subject over time.
[00072] In another aspect, there is provided herein a method for treating a
gastric cancer in
a subject, the method comprising: gastric cancer receptor agonist.
[00073] In certain embodiments, the receptor agonist is an antisense inhibitor
of one or
more of: miR-106b, miR-93 and miR-25.
[00074] In another aspect, there is provided herein a use, to manufacture a
drug for the
treatment of gastric cancer, comprised of a nucleic acid molecule chosen from
among the miR
shown in Tables 13, 14 and 16, a sequence derived therefrom, a complementary
sequence
from such miR and a sequence derived from such a complementary sequence.

8


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[00075] In certain embodiments, the drug comprises a nucleic acid molecule
presenting a
sequence chosen from among: miRs listed in Tables 13, 14 and 16, a sequence
derived from
such miRs, the complementary sequence of such miRs, and a sequence derived
from such a
complementary sequence.
[00076] In another aspect, there is provided herein an in vitro method to
identify effective
therapeutic agents or combinations of therapeutic agents to induce the
differentiation of
gastric cancer cells, the method comprising the stages of: i) culturing of
cells derived from a
gastric tumor, ii) adding at least one compound to the culture medium of the
cell line, iii)
analyzing the evolution of the level of expression of at least one miR between
stages (i) and
(ii), and iv) identifying compounds or combinations of compounds inducing a
change in the
level of expression of the miR between stages (i) and (ii).
[00077] In certain embodiments, stage (iii) includes the analysis of the level
of expression
of at least one miR.
[00078] In certain embodiments, stage (iv) includes the identification of the
compounds or
combinations of compounds modulating the level of expression of at least one
miR.
[00079] In certain embodiments, stage (iv) includes the identification of
compounds or
combinations of compounds reducing the level of expression of at least one
miR.
[00080] In certain embodiments, the compound is a therapeutic agent for the
treatment of
cancer.
[00081] Various objects and advantages of this invention will become apparent
to those
skilled in the art from the following detailed description of the preferred
embodiment, when
read in light of the accompanying drawings.

BRIEF DESCRIPTION OF THE DRAWINGS
[00082] The patent or application file may contain one or more drawings
executed in color
and/or one or more photographs. Copies of this patent or patent application
publication with
color drawing(s) and/or photograph(s) will be provided by the Patent Office
upon request and
payment of the necessary fee.
[00083] Figures 1A-1E: Alteration of miRNA expression in chronic gastritis and
gastric
adenocarcinoma. MiRNAs significantly associated with either chronic gastritis
(Fig. 1A) or
gastric adenocarcinoma (Fig. 1B) by SAM analysis (FDR=O%, q=0). Red and Green
colors
indicate upregulation and downregulation, respectively. Representative
histological features
of normal gastric mucosa, chronic gastritis and gastric adenocarcinoma are
shown,

9


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
Hematoxylin & Eosin (H&E) staining. Fig. 1C: Schematic representation of the
miR-106b-
25 cluster genomic locus hosted in the intron 13 of Mcm7. The primary
transcript of this gene
contains all the three miRNAs fused into a unique molecule that we
retrotranscribed,
amplified and sequenced from Snu-16 cells using two different sets of primers
(#1 and #2).
This molecule is not just a by-product of Mcm7 transcription as downregulation
of Drosha by
RNAi (Fig. 1D) induced a dramatic accumulation of this transcript (Fig. 1E)
confirming the
presence of an active preliminary miRNA. Bars indicate RNA expression
normalized to U6
+/- SD. This cluster shares a high degree of homology with miR-17-92 and miR-
106a-92
clusters, located on chromosomes 13 and X, respectively. Colors identify
miRNAs of the
same family.
[00084] Figures 2A-2G: E2F1 regulates miR-106b-25 expression. Fig. 2A: FACS
analysis of AGS cells synchronized in mitosis by nocodazole treatment for 12
hours and
subsequently released in fresh medium. Cells were harvested at different time
points and
analyzed for E2F1 protein content by Western Blot (Fig. 2B) and Mcm7, miR-
106b, miR-93
and miR-25 precursors RNA levels by qRT-PCR (Fig. 2C). Each analysis was
performed in
triplicate. Bars indicate RNA expression normalized to U6 +/- SD. AGS cells
were plated at
90% confluence and starved in 0.5% FBS RPMI 1640 medium for 36 hours. Cells
were then
infected with either adeno-GFP or adeno-E2F1 viruses at a M.O.I. of 25 and
incubated for
additional 21 hours: at this time, cells displayed no signs of apoptosis, as
determined by
morphology, trypan-blue staining and analysis of subdiploid DNA content (data
not shown).
MiR-106b, miR-93 and miR-25 precursors were measured by qRT-PCR as above. Snu-
16
cells were transfected with a siRNA against E2F1 (100 nM) and expression of
miR-106b-25
precursor (Fig. 2E) and mature (Fig. 2F) species was determined after 72 hours
by qRT-PCR,
as above. Bars indicate RNA expression normalized to U6 +/- SD. Fig. 2G:
Expression of
E2F1 protein in the same gastric primary tumors presented in Figure 9. Red
circles indicate
overexpression of Mcm7 and miR-106b-25 precursor RNA in the corresponding
tumors, as
determined by qRT-PCR.
[00085] Figures 3A-3F: E2F1 is a target of miR-106b and miR-93. Fig. 3A:
Endogenous
expression of mature miR-106b, miR-93 and miR-25 in human gastric cancer cell
lines and
normal mucosa determined by stem-loop qRT-PCR; bars indicate RNA expression
normalized
to U6 +/- SD. Snu-1 cells are thought to derive from a gastric neuroendocrine
tumor (NET)
while RFI and RF48 cells are from a B-cell lymphoma of the stomach. All the
other cell lines
are from gastric adenocarcinoma. Fig. 3B: Western Blot of Snu-16 cells 48
hours after



CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
inhibition of miR-106b and miR-93 by ASO transfection or (Fig. 3C)
overexpression of the
same miRNAs by oligonucleotide transfection or (Fig. 3D) lentiviral
transduction. Scramble
RNA or LNA oligonucleotides were used as negative control. Protein expression
was
quantified and normalized to GAPDH. Similar results were obtained in AGS and
MKN-74
cells (data not shown). (Fig. 3E) Luciferase assay showing decreased
luciferase activity in
cells cotransfected with pGL3-E2F1-3'UTR and miR-106b or miR-93
oligonucleotides.
Deletion of the first three bases in three putative miR- 106b/miR-93 binding
sites,
complementary to miRNA seed regions, abrogates this effect (MUT) . Bars
indicate Firefly
luciferase activity normalized to Renilla luciferase activity +/- SD. Each
reporter plasmid was
transfected at least twice (on different days) and each sample was assayed in
triplicate. Fig.
3F: qRT-PCR analysis showing E2F1 mRNA downregulation in the same cells
presented in
Fig. 3C. Bars indicate RNA expression normalized to U6 +/- SD.
[00086] Figures 4A-4E: miR-106b and miR-93 repress p21 protein expression.
Fig. 4A:
P21 expression in Snu-16 cells grown in 0.5% FBS RPMI 1640 after transfection
with either
miR-106b and miR-93 ASOs (Fig. 4A) or mimics (Fig. 4BA) or upon lentiviral
transduction
of the same miRNAs (Fig. 4C). Fig. 4D: qRT-PCR results showing no significant
difference
in p21 mRNA levels in Snu-16 cells transfected with either miR-106b or miR-93
oligonucleotides. Bars indicate RNA expression normalized to U6 +/- SD. Fig.
4E: Reporter
assay showing decreased luciferase activity in cells cotransfected with pGL3-
p21-3'UTR and
miR-106b or miR-93 oligonucleotides. Deletion of the first 3 bases of miR-
106b/miR-93
predicted binding site, complementary to miRNA seed regions, abrogates this
effect (MUT).
Bars indicate Firefly luciferase activity normalized to Renilla luciferase
activity +/- SD. Each
reporter plasmid was transfected at least twice (on different days) and each
sample was
assayed in triplicate.
[00087] Figure 5A-5D: Overexpression of miR-106b and miR-93 interfere with
TGFE-
dependent G1/S cell cycle arrest. Fig. 5A: Physiological response of Snu-16
cells to 1 ng/ml
TGFE: in the early phases of stimulation (16 hours) cells undergo a G1/S cell
cycle arrest
while apoptosis is still limited, as determined by sub-diploid DNA content.
The number of
cells undergoing apoptosis progressively increases in the following hours.
Fig. 5B:
Downregulation of E2F1 protein and (Fig. 5C) Mcm7, miR-106b, miR-93 and miR25
precursors 16 hours after TGFE stimulation. Bars indicate RNA expression
normalized to U6
+/- SD. Fig. 5D: Snu-16 cells were transfected with the indicated
oligonucleotides and
treated with 1 ng/ml TGFE after 12 hours. Upper panel: p21 protein expression.
Bottom

11


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
panel: FACS analysis, comparison of G1/S fractions between mock and miRNA
transfected
cells using unpaired t-test.
[00088] Figures 6A-6F: Inhibition of endogenous miR-106b and miR-93 expression
enhances TGFE-dependent G1/S cell cycle arrest. Fig. 6A: Analysis of cell
cycle in Snu-16
cells treated with TGFE upon inhibition of endogenous miRNAs by ASO
transfection. p-
value was calculated comparing the G1 fraction in ASOs transfected cells vs
mock-transfected
cells (unpaired t-test) (Fig. 6B) Dose-response curve of Snu-16 treated with
graded doses of
TGFP ranging from 0.1 to 5.0 ng/ml. Inhibition of endogenous miR-106b or miR-
93 by ASO
transfection restores sensitivity of Snu-16 cells to TGFP doses to which they
are otherwise
resistant (0.1 - 0.3 ng/ml), as determined by FACS analysis. * indicates
p<0.0001 (Fig. 6C).
Analysis of p21 protein and (Fig. 6D) p21 mRNA expression by Western Blot and
qRT-PCR,
respectively. Bars indicate RNA expression normalized to U6 +/- SD. The degree
of p21
protein upregulation induced by inhibition of endogenous miR-106b and miR-93
is greatly
enhanced by the presence of TGFP, possibly supported by the increased
transcription of p21
mRNA. (Fig. 6E) Snu-16 cells were transfected with a siRNA against p21 alone
or in
combination with either miR-106b or miR-93 mimics and treated with 1 ng/ml
TGFP for 16
hours. While miR-106b lost all of its effect on cell cycle, miR-93 still
maintained a residual
effect after p21 silencing. This differential response between miR-106b and
miR-93 is
statistically significant (p=0.0272) (Fig. 6F) Analysis of expression by
Western Blot of
various proteins involved in the G1/S checkpoint upon TGFP stimulation.
[00089] Figures 7A-7G: miR-25 cooperates with miR-106b and miR-93 in
preventing the
onset of TGFP-induced apoptosis CCK-8 viability assay of Snu-16 cells
transfected with
miRNA mimics. * indicates significant difference (p<0.001) in the number of
viable cells
upon transfection of miR-106b, miR-93, miR-25 and/or miR-106b-25 and
subsequently
treated with 1 ng/ml TGFP for 48 hours. (Fig. 7B) Conversely, inhibition of
miR-106b, miR-
93 and miR-25 cooperatively augments the response to TGFP: statistical
significance
(p<0.001) was reached upon transfection of a mixture of the three ASOs. (Fig.
7C)
Significant loss of viability was confirmed by analysis of subdiploid DNA
content. (Fig. 7D)
Bim protein expression in Snu-16 cells at 48 hours post-transfection with
either miRNA
mimics or ASOs or after lentiviral transduction of the same miRNAs. Same
effects on Bim
expression were obtained in AGS and MKN-74 cells (data not shown). (Fig. 7E)
Luciferase
assay showing decreased luciferase activity in cells cotransfected with pGL3-
Bim-3'UTR and
miR-25. Deletion of the first 3 bases of miR-25 predicted binding sites,
complementary to

12


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
miRNA seed regions, abrogates this effect (MUT). Bars indicate Firefly
luciferase activity
normalized to Renilla luciferase activity +/SD. Each reporter plasmid was
transfected at least
twice (on different days) and each sample was assayed in triplicate. (Fig. 7F)
qRT-PCR
analysis showing no difference in Bim mRNA (Taqman probe recognizing the two
major
isoforms Bim EL and Bim L) in Snu-16 cells transfected with miR-25
oligonucleotide. Bars
indicate RNA expression normalized to U6 +/- SD. (Fig. 7G) FACS analysis of
subdiploid
DNA content in Snu-16 cells transfected with miR-25 oligonucleotide, si-Bim,
both or a
scramble oligonucleotide and subsequently treated with 1 ng/ml TGFEO. for 24
hours.
Statistical analysis as above.
[00090] Figure 8: The E2F1/miR-106b-25/p21 pathway. A model summarizing the
mechanism of action of miR-106, miR-93 and miR-25 described herein.
[00091] Figure 9A-9D: Expression of the miR-106b-25 cluster in gastric cancer.
Fig. 9A:
293T/17 cells were transfected with 100 nM miRNA oligonucleotides (Ambion), as
indicated,
and assayed for miRNA expression by stem-loop qRT-PCR. MiR-106b, miR-93, miR-
25
primers showed high specificity while miR-17-5p and miR-92 primers cross-
hybridized with
miR-106a and miR-25, respectively. Results were normalized to U6 and converted
to the
same scale. Expression of mature (Fig. 9B) and precursor (Fig. 9C) miRNAs in a
set of 10
gastric primary tumors and 10 non-tumor controls, as determined by qRT-PCR.
Bars
represent relative fold-changes between tumor and non-tumor tissues from the
same patient
+/- SD. Each sample was analyzed in triplicate and normalized to either RNU49
(mature
miRNAs) or CAPN2 (precursor miRNAs and Mcm7): these genes showed the least
variability
(<0.4 Ct values) among 12 different normalizers tested in these samples. (Fig.
9) Snu-16 cells
were transduced with a lentiviral vector carrying miR- 106b, miR-93, miR-25 or
the miR-
106b-25 cluster and mature miRNA levels were measured after 72 hours by qRT-
PCR. Bars
indicate RNA expression normalized to U6 +/- SD and converted to the same
scale.
Transduction efficiency >90% was confirmed by fluorescent microscopy. Similar
results
were obtained in AGS cells (data not shown).
[00092] Figures 1OA-10G: Proliferation studies in cells with high/low miR-106b-
25 (basal
conditions). FACS analysis and proliferation curves of Snu-16 (Fig. 10A, Fig.
10C) and AGS
(Fig. 10B, Fig. 10D) cells transfected with miRNA ASOs or mimics,
respectively. (Fig. 10E)
Proliferation curves of AGS cells stably transduced with a fluorescent
lentiviral vector
carrying miR-106b, miR-93, miR-25 or miR-106b-25 precursors under control of a
CMV
promoter. Infection efficiency > 95% was determined by fluorescent microscopy.
(Fig. 10F)
13


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
Colony formation assay: AGS cells were transfected with pRetroSuper-Puro
constructs
encoding miR-106b, miR-93, miR-25 or miR-106b-25 precursors or a scramble
sequence and
grown in 2 ug/ml puromycin for 14 days. Efficient miRNA expression and
processing by all
these constructs were assayed by Northern Blot and stem-loop qRT-PCR (data not
shown).
(Fig. 10G) Proliferation curve and (H) FACS analysis of Snu-16 cells in which
either p21 or
E2F1 were selectively silenced by RNAi. Inhibition of p21 expression produced
an effect on
cell cycle that was undistinguishable from miR-93.
[00093] Figure 11: MKN-74 cell viability assay in the presence of TGFE. MKN-74
cells
were transfected with the indicated LNA oligonucleotides to silence endogenous
miRNA
expression and subsequently treated with TGFE for 96 hours. Cell viability was
determined
by CCK-8 assay.
[00094] Figure 12: Annexin V assay of miR-25 overexpressing cells. Results
shown in
Figure 7G were confirmed by Annexin V staining.
[00095] Figure 13: Table 1: Differentially expressed miRNAs in chronic
gastritis VS
normal gastric mucosa.
[00096] Figure 14: Table 2: Differentially expressed miRNAs in gastric
adenocarcinomas
VS non-tumor gastric mucosa.
[00097] Figure 15: Table 3: MicroRNA expression in human gastric cancer cell
lines.
[00098] Figure 16: Table 4: Validation of microarray data in paired human
primary
tumors VS non-tumor controls by qRT-PCR.
[00099] Figure 17: Table 5: Mcm7 mRNA and miR-106b-25 expression in 10 paired
gastric primary tumors and non-tumor controls.
[000100] Figure 18: Table 6: Human genes harboring putative miR-106b, miR-93
and miR-
25 binding sites on the same 3' UTR.

DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
[000101] Throughout this disclosure, various publications, patents and
published patent
specifications are referenced by an identifying citation. The disclosures of
these publications,
patents and published patent specifications are hereby incorporated by
reference into the
present disclosure to more fully describe the state of the art to which this
invention pertains.
[000102] Deregulation of E2F1 activity and resistance to TGFE are hallmarks of
gastric
cancer. MicroRNAs (miRNAs) are small non-coding RNAs frequently misregulated
in
human malignancies.

14


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000103] Here we show that miR-106b-25 cluster, upregulated in a subset of
human gastric
tumors, is activated by E2F1 in parallel with its host gene Mcm7. In turn, miR-
106b and miR-
93 regulate E2F1 expression, establishing a miRNA-directed negative feedback
loop.
Furthermore, upregulation of these miRNAs impairs the TGFE tumor suppressor
pathway
interfering with the expression of CDKNIA (p2lWafl/Cipl) and BCL2L11 (Bim).
Together,
these results show that miR-106b-25 cluster is involved in E2F1 post-
transcriptional
regulation and can play a key role in the development of TGFE resistance in
gastric cancer.
[000104] MicroRNAs (miRNAs) are small non-coding RNAs that may regulate the
expression of approximately 30% of all human genes, either inhibiting target
mRNA
translation or inducing its degradation. These genes are abnormally expressed
in human
malignancies, making their biological importance increasingly apparent.
Gastric cancer
causes 12% of all cancer-related deaths each year calling for better
treatments based on a
deeper understanding of the molecular mechanisms underlying the onset of this
disease.
[000105] Here, we show that overexpression of the miR-106b-25 cluster leads to
deregulation of important cancer-related genes, such as the TGFE effectors
p21Wafl/Cipl
and Bim, disrupting the G1/S checkpoint and conferring resistance to TGFE-
dependent
apoptosis.
[000106] We also show that microRNAs (miRNAs) may be involved in gastric
tumorigenesis. MiRNAs are non-protein coding genes thought to regulate the
expression of
up to 30% of human genes, either inhibiting mRNA translation or inducing its
degradation
(Lewis et al., 2005). Besides a crucial role in cellular differentiation and
organism
development (Kloosterman and Plasterk, 2006), miRNAs are frequently
misregulated in
human cancer (Lu et al., 2005; Volinia et al., 2006) and they can act as
either potent
oncogenes or tumor suppressor genes (Esquela-Kersher et al. 2006).
[000107] Here we show that E2F1 regulates miR-106b, miR-93 and miR-25, a
cluster of
intronic miRNAs hosted in the Mcm7 gene, inducing their accumulation in
gastric primary
tumors. Conversely, miR-106b and miR-93 control E2F1 expression, establishing
a negative
feedback loop that may be important in preventing E2F1 self-activation and,
possibly,
apoptosis.
[000108] On the other hand, we found that miR-106b, miR-93 and miR-25
overexpression
causes a decreased response of gastric cancer cells to TGFE interfering with
the synthesis of
p21 and Bim, the two most downstream effectors of TGFE-dependent cell cycle
arrest and
apoptosis, respectively.



CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000109] These miRNAs contribute to the onset of TGFE resistance in cancer
cells and now
believed by the inventors herein to represent novel therapeutic targets for
the treatment of
gastric cancer.
[000110] The present invention is further explained in the following Examples,
in which all
parts and percentages are by weight and degrees are Celsius, unless otherwise
stated. It
should be understood that these Examples, while indicating preferred
embodiments of the
invention, are given by way of illustration only. From the above discussion
and these
Examples, one skilled in the art can ascertain the essential characteristics
of this invention,
and without departing from the spirit and scope thereof, can make various
changes and
modifications of the invention to adapt it to various usages and conditions.
All publications,
including patents and non-patent literature, referred to in this specification
are expressly
incorporated by reference.
[000111] EXAMPLE I
[000112] Deregulation of miRNA expression in human gastric cancer
[000113] Most gastric adenocarcinomas arise in the context of a chronic
inflammatory
background, frequently associated with Helicobacter Pylori (HP) infection
(Uemura et al.,
2001). Nevertheless, the molecular mechanisms responsible for HP oncogenicity
are poorly
understood, although Th1 immune response seems to be critical in the
development of
preneoplastic lesions such as gastric atrophy and intestinal metaplasia
(Houghton et al., 2002;
Fox et al., 2000).
[000114] In the search of miRNAs potentially involved in gastric
tumorigenesis, we
analyzed global miRNA expression in 20 gastric primary tumors of the
intestinal-type, each
one paired with adjacent nontumor gastric tissue from the same patient, and 6
gastric cancer
cell lines using a custom miRNA microarray. To identify specific alterations
associated with
inflammation and/or preneoplastic lesions, we first compared non-tumor tissues
with
histological signs of chronic gastritis (n=13) versus otherwise normal mucosa
(n=7). Seven
miRNAs were associated with chronic inflammation by unpaired Significance
Analysis of
Microarrays (SAM), including miR-155 that is known to predispose to cancer
(Costinean et
al., 2006) and to play a major role in the regulation of immune response
(Rodriguez et al.,
2007; Thai et al., 2007) (Figure 1A, Figure 13 - Table 1).
[000115] We then examined the miRNA expression profile of gastric primary
tumors and
cancer cell lines: a total of 14 miRNAs exhibited a 2-fold or greater median
overexpression in
primary tumors compared to non-tumor controls by paired SAM (Figure 1B, Figure
14 -

16


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
Table 2). Of these, 13 out of 14 ranked above the 80th percentile in all
gastric cancer cell
lines in terms of expression, except for miR-223 that was not expressed
(Figure 15 - Table 3).
Only 5 miRNAs were downregulated in cancer (Figure 1B, Figure 14 - Table 2).
Microarray
data were confirmed by stem-loop qRT-PCR for 9 out of 10 tested miRNAs (Figure
16 -
Table 4). Among the misregulated miRNAs, miR-21, miR-223, miR-25 and miR-17-5p
showed the highest overexpression in tumors, with 4.5, 4.2, 3.7 and 3.7 median
fold-changes,
respectively.
[000116] These results indicate that specific modifications in the miRNA
expression pattern
are characteristic of human gastric cancer since the earliest steps of
tumorigenesis and involve
miRNAs with known oncogenic properties, such as miR-21 (Meng et al., 2006) and
miR-17-
5p (He et al., 2005).
[000117] miR-106b-25 cluster is overexpressed in gastric cancer
[000118] Among the overexpressed miRNAs, miR-25 were discovered to be
especially
useful an attractive candidate for playing a role in gastric tumorigenesis. In
fact, this was the
3rd most strongly upregulated miRNA in primary gastric tumors (median fold-
change: 3.7;
range 1.0 - 26.8) and ranked among the most highly expressed miRNAs in all
human gastric
cancer cell lines (above 97th percentile). miR-106b (median fold-change: 2.0;
range 1.0 - 6.5)
and miR-93 (median fold-change: 2.3; range 1.0 - 7.7) were also upregulated in
primary
tumors and highly expressed in all gastric cancer cell lines (above 82nd and
89th percentile,
respectively).
[000119] These three miRNAs (hereafter miR-106b-25) are clustered in the
intron 13 of
Mcm7 on chromosome 7q22 and actively cotranscribed in the context of Mcm7
primary RNA
transcript (Kim et al., 2007 and Figure 1C-E). Several studies reported the
amplification of
this region in gastric tumors (Weiss et al., 2004; Peng et al., 2003; Takada
et al., 2005).
However, we could not detect any amplifications of the miR-106b-25 locus in
our samples
(data not shown), implying that other mechanisms must contribute to miR-106b-
25
overexpression in gastric cancer.
[000120] Mcm7 plays a pivotal role in the G1/S phase transition, orchestrating
the correct
assembly of replication forks on chromosomal DNA and ensuring that all the
genome is
replicated once and not more than once at each cell cycle (Blow and Hodgson,
2002). As
overexpression of Mcm7 has been associated with bad prognosis in prostate and
endometrial
cancer (Ren et al., 2006; Li et al., 2005) we hypothesized that Mcm7
oncogenicity may be
linked, at least in part, to overexpression of the hosted miRNAs. Moreover,
the miR-106b-25

17


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
cluster shares a high degree of homology with the miR-17-92 cluster (Figure
1C), which
appears to have an oncogenic role (He et al., 2005; O'Donnell et al., 2005;
Dews et al., 2006).
[000121] We then investigated the miR-106b-25 cluster. We first determined the
specificity
of stem-loop qRT-PCR. Primers for miR-106b, miR-93 and miR-25 were highly
specific
while miR-17-5p and miR-92 probes cross-hybridized with miR-106a and miR-25,
respectively (Figure 9A). Next, we used stem-loop qRT-PCR to assay the
expression of
mature miRNA species in an independent set of ten gastric primary tumors
paired with non-
tumor gastric mucosa from the same patient.
[000122] Mature miR-106b, miR-93 and miR-25 were overexpressed in 6/10, 6/10
and 5/10
of these tumors, respectively, although there was not reciprocal correlation
in their level of
expression (Figure 9B).
[000123] We examined miRNA precursor levels in the same tumors by conventional
qRT-
PCR (Figure 9C) and we found miR-106b, miR-93 and miR-25 precursor species to
be
concordantly expressed in the tumors [r(106b/93)=0.93; r(106b/25)=0.78;
r(93/25)=0.88,
Figure 17 - Table 5].
[000124] Of the 5 tumors overexpressing miR-106b-25 precursors, 3 tumors also
expressed
high levels of mature miR-106b, miR-93 and miR-25 whereas the remaining tumors
displayed
variable expression of each mature miRNA, showing an additional level of post-
transcriptional regulation controlling individual miRNAs.
[000125] Mcm7 mRNA was also overexpressed in 5/10 tumors, showing an almost
perfect
correlation with miR-106b, miR-93 and miR-25 precursor levels (r=0.98, 0.92,
0.72,
respectively, Figure 9C and Figure 17 - Table 5).
[000126] These data show that miR-106b-25 precursors are specifically
overexpressed in a
subset of gastric primary tumors in parallel with Mcm7 mRNA. Although we
cannot exclude
the possibility of a miR-106b-25 independent promoter, our results show that
miR-106b-25
transcription in gastric tumors is driven by its host gene Mcm7. Moreover, a
post-
transcriptional mechanism also plays a major role in determining the levels of
mature miR-
106b-25, as recently proposed for other miRNAs (Thomson et al., 2006).
[000127] A negative feedback loop controls E2F1 and miR-106b-25 expression.
[000128] E2F1 is a transcription factor that transactivates a variety of genes
involved in
chromosomal DNA replication (Johnson and DeGregori, 2006), including Mcm7
(Suzuki et
al., 1998; Arata et al., 2000). The inventors herein now believe that miR-106b-
25
transcription may be similarly regulated by E2F1. To test, we first determined
whether

18


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
endogenous fluctuations in E2F1 protein levels corresponded to similar changes
in Mcm7 and
miR-106b-25 expression. Interestingly, AGS gastric cancer cells, arrested in
mitosis by
nocodazole treatment for 12 hours did not express E2F1 protein and showed
reduction in
Mcm7 transcript (2-fold) and miR-106b, miR-93 and miR-25 precursors (4.0, 5.2
and 12.0-
fold, respectively), compared to exponentially growing cells. As cells were
released and re-
entered the G1 phase, E2F1 expression paralleled Mcm7, miR-106b, miR-93 and
miR-25
precursor RNA reaccumulation. (Figures 2A-C).
[000129] This process was directly associated with E2F1 expression because its
specific
overexpression by adenoviral transduction (Figure 2D) or silencing by RNA
interference
(Figure 2E) also induced consistent changes in miR-106b-25 precursor levels.
E2F1 loss of
function impacted the expression of mature miRNAs after 72 hours, as well
(Figure 2F).
[000130] To further validate the data in vivo, we analyzed E2F1 protein
expression in 10
primary gastric tumors by Western Blot and found a positive correlation
between E2F1
protein and Mcm7/miR-106b-25 precursor expression (Figure 2G). In fact, 4 out
of 5 tumors
overexpressing E2F1 displayed higher levels of Mcm7 and miR-106b-25 precursors
(Figure
9C). Of these, 3 tumors also overexpressed mature miR-106b, miR-93 and miR-25
(Figure
9B). However, one tumor showed Mcm7 and miR-106b-25 precursors upregulation
without
detectable levels of E2F1, showing that other transcription factors are also
involved in the
regulation of miR-106b-25.
[000131] These results indicate that E2F1 regulates miR-106b-25 expression in
parallel with
Mcm7, showing that overexpression of these miRNAs in gastric cancer is due, at
least in part,
to E2F1 upregulation.
[000132] Recently, miR-17-5p has been proposed as a novel post-transcriptional
regulator of
E2F1 (O'Donnell et al., 2005). Given the similarity between miR-17-5p, miR-
106b and miR-
93 sequences, we explored the possibility that also miR-106b and miR-93 may
participate in
the regulation of E2F1 expression. Because these miRNAs were diffusely
expressed in a
panel of 12 gastric cancer cell lines analyzed by qRT-PCR (Figure 3A) we
adopted a loss of
function approach to antagonize miR-106b-25. Transfection of LNA antisense
oligonucleotides (ASOs) against miR-106b and miR-93 induced an accumulation of
E2F1
protein in Snu-16 cells indicating that endogenous levels of these miRNAs
control its
expression (Figure 3B).
[000133] Also, overexpression of these miRNAs by either oligonucleotide
transfection or
lentiviral transduction (Figure 9D) clearly decreased E2F1 protein levels in
Snu- 16 and AGS
19


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
gastric cancer cell lines (Figure 3C, and Figure 3D) and inhibited the
expression of a reporter
vector containing E2F1 3'UTR. Mutation of the predicted miRNA binding sites in
the
reporter vector abrogated this effect indicating that miR- 106b and miR-93
directly interact
with E2F1 3'UTR (Figure 3E). However, E2F1 mRNA decreased by 2-fold upon miR-
106b
and miR-93 transfection, possibly because of partial mRNA degradation or
downmodulation
of E2F1 transcriptional activators (Figure 3F).
[000134] It has been argued that miR-17-5p may secondarily inhibit E2F1
expression by
suppressing AIB-1 protein that in fact activates E2F1 transcription and is
also a miR-17-5p
target (Hossain et al., 2006). While it is very reasonable that miRNAs act on
different targets
within the same pathway, we analyzed AIB-1 protein levels in AGS and Snu-16
cells and we
found a slight decrease or no difference at all in cells transfected with
either miR- 106b or
miR-93, respectively, showing that AIB-1 is a bona fide low affinity target of
miR-106b that
may only partially contribute to E2F1 downregulation (Figure 3C).
[000135] Together these results show that E2F1 regulates miR-106b-25
expression but is
also a target of miR-106b and miR-93, establishing a negative feedback loop in
gastric cancer
cells. Because E2F1 is known to self-activate its own promoter through a
positive feedback
loop these miRNAs may control the rate of E2F1 protein synthesis preventing
its excessive
accumulation, as recently proposed for homolog miR-17-5p and miR-20a
(Sylvestre et al.,
2007; Woods et al., 2007).
[000136] miR-106b and miR-93 impair TGFE-induced cell cycle arrest.
[000137] These results show that miR-106b-25 transcription is promptly induced
by E2F1 as
cells exit mitosis and re-enter the G1 phase. On this basis, we hypothesized a
possible role for
miR-106b-25 in repressing GO/G1 associated activities, ideally cooperating
with E2F1. So,
we interrogated TargetScan database looking for genes known to be negatively
regulated by
E2F1 and we identified CDKNIA (p21) as a putative target of miR-106b and miR-
93. This
gene, frequently dysfunctional in human cancer, is a key inhibitor of the cell
cycle (Mattioli et
al, 2007). Intriguingly, we confirmed that miR-106b and miR-93 endogenously
expressed in
Snu-16 cells post-transcriptionally regulate p21. In fact, their inhibition by
ASOs enhanced
the expression of p21 protein (Figure 4A). Conversely, upregulation of miR-
106b and miR-
93 achieved by either oligonucleotide transfection (Figure 4B) or lentiviral
transduction
(Figure 4C) repressed p21 protein expression without significant changes in
p21 mRNA
levels (Figure 4D). Moreover, miR-106b and miR-93 mimics inhibited the
expression of a
reporter vector containing p21 3' UTR while mutation of the predicted miRNA
binding site



CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
abrogated this effect (Figure 4E).
[000138] Given the importance of p21 in the regulation of cell cycle, we
decided to address
the role of miR106b-25 in controlling the proliferation of gastric cancer
cells. Unexpectedly,
loss of miR-106b, miR-93 and/or miR-25 function induced by ASOs transfection
did not
produce any significant alterations in the cell cycle and proliferation of Snu-
16 cells (Figure
10A and Figure 10C). Similarly, overexpression of the three miRNAs by either
oligonucleotide transfection or lentiviral transduction did not significantly
modify the
proliferation rate and colony formation efficiency of AGS cells, although we
noticed limited
but reproducible cell cycle perturbations upon miR-93 overexpression (+8% of
cells in S
phase, Figures 10B, 10D and 10E).
[000139] We obtained similar results using GTL-16 and MKN-74 gastric cancer
cell lines
(data not shown) indicating that miR-106b-25 function is not essential for the
survival and the
proliferation of gastric cancer cells in vitro. However, specific silencing of
either p21 or E2F1
by RNAi produced no significant alterations in the proliferation as well
(Figures 1OG and
10H), confirming that these cancer cell lines are not responsive to p21 basal
levels and can
well compensate for the loss of E2F1 expression.
[000140] We then addressed the role of miR-106b-25 in the presence of TGFO:
this
cytokine, by inducing the expression of p21 and other antiproliferative
molecules, ensures
timely coordinated cell cycle arrest and apoptosis of mature cells in the
gastrointestinal tract,
thus controlling the physiological turnover of epithelial cells (van den Brink
and Offerhaus,
2007). Impairment of this crucial tumor suppressor pathway is a hallmark of
gastric cancer
(Ju et al., 2003; Park et al., 1994). However, Snu-16 cells are among the few
gastric cancer
cell lines still responding to relatively high doses of TGFO in vitro,
undergoing G1/S arrest
and subsequent massive apoptosis (Ohgushi et al., 2005 and Figure 5A).
Nevertheless, cell
viability decreases after 24 hours, this opening a window to study early
molecular changes
associated with TGFO.
[000141] Interestingly, stimulation with TGFO induced marked downregulation of
E2F1
protein, Mcm7 mRNA and miR-106b-25 precursors after 16 hours, when cells
physiologically
undergo G1/S arrest, suggesting that downmodulation of these miRNAs is part of
the
physiological response to TGFO (Figures 5B and 5C). To establish the
importance of this
process, we counteracted miR-106b-25 downregulation by introducing miR-106b,
miR-93
and/or miR-25 mimics in Snu-16 cells in the presence of TGFO. Notably,
overexpression of
miR-93 completely abrogated TGFO-induced cell cycle arrest while miR-106b
partially

21


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
decreased it (p<0.0002), consistent with the degree of p21 downregulation
induced by these
miRNAs (Figure 5D).
[000142] Conversely, antagonizing endogenous miR-106b and miR-93 expression by
ASOs
significantly increased the number of Snu-16 cells undergoing TGFO-dependent
cell cycle
arrest (p<0.0013) and restored sensitivity to suboptimal doses of TGFO
(p<0.0001) to which
these cells are otherwise resistant (Figures 6A and 6B).
[000143] Accordingly, the degree of p21 upregulation achieved by inhibiting
endogenous
miR106b and miR-93 in the presence of TGFO (Figure 6C) was double than in
basal
conditions (Figure 4A), probably supported by the active transcription of p21
mRNA (Figure
6D).
[000144] To establish the role of p21 in inducing the phenotype associated
with miR-106b
and miR-93 gain/loss of function, we specifically silenced p21 by RNAi (si-
p21) in Snu-16
cells treated with TGFP. This recapitulated almost in full the effect of miR-
106b and miR-93
overexpression on cell cycle distribution (Figure 5D) whereas cotransfection
of si-p21 with
miR-106b and miR-93 dramatically reduced the effect of these miRNAs on TGFO-
induced
cell cycle arrest, suggesting that p21 is a primary target in this biological
context (Figure 6E).
However, a small but statistically significant effect on TGFO-dependent cell
cycle arrest by
miR-93 was still observable in the absence of p21 (p=0.0272), implying that
other direct or
indirect targets cooperate with p21. Analysis of expression for genes involved
in the G1/S
checkpoint point at p27 as a possible indirect target of miR-93 (Figure 6F).
[000145] These data show that miR-106b and miR-93 interfere with TGFO-induced
cell
cycle arrest mainly inhibiting the expression of p21 at the post-
transcriptional level. However,
p21-independent pathways may be also involved in delivering the complete
effect of miR-93
on cell cycle control.
[000146] miR-25 cooperates with miR-106b and miR-93 in preventing the onset of
TGFO-
induced apoptosis.
[000147] These results show a role for miR-106b and miR-93 in modulating the
cell cycle
arrest in the early phase of TGFP stimulation. We analyzed miR-106b-25
function upon
prolonged exposure to TGFP that eventually results in apoptosis (Ohgushi et
al., 2005, and
Figure 5B).
[000148] We examined the viability of Snu-16 cells stimulated with TGFP for 24-
48 hours
by tetrazolium reduction assay. Introduction of miR-106b, miR-93 and/or miR-25
mimics in
these cells induced marked resistance to TGFP (Figure 7A). Conversely, ASOs
transfection
22


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
induced a negative trend in the number of viable cells that reached
statistical significance
(p=0.003) when all the three miRNAs were inhibited at the same time (Figure
7B). This
result was confirmed by FACS analysis that showed a significant increase in
the number of
subdiploid cells upon silencing of the three miRNAs (p<0.001). Moreover, the
higher
sensitivity of this assay allowed detection of smaller but significant changes
(p<0.001) in the
percentage of subdiploid cells upon individual inhibition of miR-106b, miR-93
or miR-25
(Figure 7C). Finally, silencing of miR-106b-25 partially restored sensitivity
to TGFE in
otherwise resistant MKN-74 cells (Figure 11). Together, these results are
consistent with a
model where endogenous miR-106b, miR-93 and miR-25 cooperate in modulating the
expression of one or more targets mediating TGFE-dependent apoptosis.
[000149] We searched TargetScan database looking for effectors of apoptosis
and we
identified BCL2L11 (Bim) as the only strong candidate out of 18 human genes
harboring
putative binding sites for miR- 106b, miR-93 and miR-25 at the same time
(Figure 18 - Table
6). Bim is a 1313-only protein that critically regulates apoptosis in a
variety of tissues by
activating proapoptotic molecules like Box and Bad and antagonizing
antiapoptotic molecules
like Bc12 and Bill (Gross et al., 1999). A fine balance in the intracellular
concentrations of
Bim and its partner proteins is crucial in order to properly regulate
apoptosis. Bim is
haploinsufficient and inactivation of even a single allele accelerates Myc-
induced
development of tumors in mice without loss of the other allele (Egle et al.,
2004). Notably,
Bim is the most downstream apoptotic effector of the TGFE pathway and its
downmodulation
abrogates TGFE-dependent apoptosis in Snu-16 cells (Ohgushi et al., 2005).
[000150] We determined whether Bim was a direct target of miR-106b-25. Snu-16
cells
express all the three major isoforms of Bim, namely Bim EL, Bim L and Bim S.
Intriguingly,
antagonizing endogenous miR-25 by ASOs transfection induced an accumulation of
all the
three isoforms in Snu-16 cells whereas miR-25 overexpression by either
oligonucleotide
transfection or lentiviral transduction reduced their expression. On the
contrary, miR-106b
and miR-93 did not influence Bim expression in 3 out of 3 tested gastric
cancer cell lines
(Figure 7D).
[000151] While it is still possible that miR-106b and miR-93 cooperate with
miR-25 in
regulating Bim expression in other tissues, this supports a model where
multiple effectors of
apoptosis are coordinately repressed by each of the three miRNAs in gastric
cancer.
[000152] We focused on Bim as one of these apoptotic effectors and determined
that miR-25
predicted binding sites on its 3'UTR mediate target recognition and subsequent
inhibition of
23


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
translation by luciferase assay (Figure 7E). Moreover, Bim EL and Bim L mRNA
levels
were unchanged in Snu-16 cells upon miR-25 overexpression, which is indicative
of a post-
transcriptional regulatory mechanism (Figure 7F).
[000153] In order to establish the importance of Bim downregulation relative
to miR-25
specific antiapoptotic function, we suppressed Bim protein in Snu-16 cells
using a siRNA
against its three major isoforms (si-Bim, Figure 7D) and we subsequently
treated these cells
with TGFE for 24 hours. Notably, protection from apoptosis conferred by si-Bim
and miR-25
was very similar, as determined by sub-diploid DNA content and Annexin V
staining.
Moreover, co-transfection of Bim and miR-25 did not have significant additive
effects
(p=0.6328), suggesting that Bim downregulation is a main mechanism of
resistance to TGFE-
induced apoptosis in miR-25 overexpressing cells (Figure 7G and Figure 12).
[000154] We show that miR-106b-25 cluster, activated by E2F1 and upregulated
in human
gastric adenocarcinomas, alters the physiological response of gastric cancer
cells to TGFE
affecting both cell cycle arrest and apoptosis (Figure 8).
[000155] These findings are of particular relevance in a gastric cancer model
as impairment
of the TGFE tumor suppressor pathway is a critical step in the development of
gastric tumors.
[000156] Discussion
[000157] We performed a genome-wide analysis of miRNA expression in different
steps of
gastric carcinogenesis. Since the vast majority of gastric tumors originate
from a chronic
inflammatory background (Uemura et al., 2001), we considered of particular
relevance
discriminating between preneoplastic and tumor-specific alterations.
[000158] For the first time, we identified the specific overexpression of a
miRNA cluster in
human tumors that had been ignored thus far. Although we focused on gastric
cancer,
overexpression of miR-106b, miR-93 and miR-25 in other types of cancer may be
a common,
yet underestimated, event.
[000159] In fact, miR-106b-25 expression is intimately linked with the
expression of E2F1
and Mcm7 that are involved in basic mechanisms of cellular proliferation. For
example,
Mcm7 is frequently overexpressed in prostate cancer (Ren et al., 2006) and, in
fact, we
previously described miR-25 upregulation in a large-scale miRNA study on this
type of cancer
(Volinia et al., 2006). Moreover, we showed that stem-loop qRT-PCR probes
commonly used
in assaying the expression of miR-92, that is overexpressed in most human
tumors (Volinia et
al., 2006), cross-hybridize with miR-25. However, given the nearly identical
sequences, it is
very likely that miR-106b-25 and miR-17-92 cooperate in exerting similar, if
not identical,

24


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
functions: in fact, we found that miR-17-5p, miR-18a and miR-20a also inhibit
p21 expression
whereas miR-92 represses Bim expression (F.P. and AN., unpublished data).
Moreover, both
miR-106b-25 and miR-17-92 are regulated by E2F1. These clusters also exhibit
some
differences, though. For example, miR-106b resembles miR-17-5p but it is three
nucleotides
shorter: it has been reported that specific sequences in the 3' termini can
define the
intracellular localization of miRNAs (Hwang et al., 2007). Moreover, the miR-
19 family is
not represented in the miR-106b-25 cluster (Figure 2A).
[000160] On the other hand, miR-93 belongs to the same family of miR-372 and
miR-373:
these miRNAs are overexpressed in testicular germ cell tumors where they
impair LATS2
expression, making cells insensitive to high p21 levels (Voorhoeve et al.,
2006).
[000161] As shown herein, miR-93 acts within the same pathway, directly
targeting p21
expression. Therefore, this family of miRNAs is now believed to be involved in
the control of
a crucial hub for the regulation of cell cycle and may have particular
relevance in cancer.
[000162] Moreover, miR-93 shares high sequence homology with miR-291-3p, miR-
294 and
miR-295: these miRNAs are specifically expressed in pluripotent ES cells and
they are either
silenced or downregulated upon differentiation (Houbaviy et al., 2003). While
not wishing to
be bound by theory, the inventors herein now believe that these miRNAs may be
similarly
involved in the regulation of p21.
[000163] The inventors show that miRNAs play a role in the control of cell
cycle through
different mechanisms. In the case of E2F1, miRNAs seem to act mainly in the
context of
regulatory, redundant feedback loops. In fact, miR-106b, miR-93, miR-17-5p and
miR-20a,
located on separate miRNA clusters, are regulated by E2F1 and presumably
cooperate in
inhibiting its translation.
[000164] At the same time, we found these miRNAs to be involved in the control
of p21
expression and early response to TGFE. The inventors also believe that they
also control
other tumor suppressor pathways converging on p21. Loss of p21 function by
mutation,
deletion, hypermethylation, ubiquination or mislocalization is a frequent
event and a negative
prognostic factor in human gastric cancer (Mattioli et al., 2007). However,
the role of
miRNAs in p21 regulation had never been explored before. Since 80% of the
studied gastric
primary tumors did not express p21 protein at detectable levels we could not
establish an
inverse correlation between miRNAs and p21 protein expression. However, p21
mRNA
levels in primary tumors were often comparable to normal tissues, indicating
post-
transcriptional regulation as a frequent cause of p21 downregulation in
gastric cancer (F.P and



CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
A.V., unpublished data).
[000165] Interestingly, induction of p21 expression seemed to be a
prerequisite to elicit a
miR-106b/miR-93 associated response in the early phase of TGFE stimulation.
Conversely,
silencing p21 by RNAi dramatically decreased the effect of these miRNAs on
cell cycle.
Although hundreds of different targets are predicted for each miRNA by
computational
methods there is increasing evidence that "primary miRNA targets" may be
critical for
specific biological functions. For example, miR-10b enhances cell motility and
invasiveness
of breast cancer cells but this phenotype is completely reverted upon
constitutive expression
of its target HOXD 10, although over one hundred targets are predicted for
this miRNA (Ma
and Weinberg, 2007). It is to be noted, of course, these observations do not
exclude other
contexts where parallel regulation of multiple targets by a single miRNA is
necessary to exert
a specific function. Furthermore, multiple miRNAs may cooperate in exerting
the same
function.
[000166] This is the case of the miR-106b-25 cluster that protects gastric
cancer cells from
apoptosis. Such effect is partitioned between the three miRNAs that cooperate
in repressing
the expression of different proapoptotic molecules. We identified Bim, the
most downstream
apoptotic effector of the TGFE pathway (Ohgushi et al., 2005), as a key target
of miR-25.
This is of particular relevance in a gastric cancer model. In fact, TGFE is
one of the main
regulators of gastric homeostasis and is essential in regulating the
physiological turnover of
epithelial cells through apoptosis (van den Brink and Offerhaus, 2007). While
the identity of
miR-106b and miR-93 proapoptotic targets remains elusive, we could clearly
detect
antiapoptotic and proapoptotic responses associated with miR-106b, miR-93
and/or miR-25
overexpression and inhibition, respectively; these properties emerge in the
late phase of TGFE
stimulation when cell cycle arrest is revoked and apoptosis becomes the
dominant process
characterizing the response of gastric cells to TGFE. The small but
significant alterations
observed upon inhibition of single miRNAs, readily detected by analysis of
subdiploid DNA
content, acquire biological consistency when the three ASOs are delivered
together,
confirming the cooperative relationship between these clustered miRNAs.
[000167] Although a negative trend was observed in TGFE-stimulated cells
transfected with
single ASOs by both tetrazolium reduction assay and analysis of subdiploid DNA
content, this
did not reach statistical significance in the tetrazolium reduction assay.
This is to be imputed
to the 5-10% standard error associated with this assay that statistically
excludes smaller
differences. On the contrary, the standard error in the analysis of subdiploid
DNA content
26


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
was below 2% in our hands.
[000168] When we looked at Bim expression in primary tumors we noticed general
overexpression compared to normal tissues (F.P. and A.V. unpublished data).
This is
consistent with previous studies showing that Bim is induced by oncogenic
stress as a
safeguard mechanism to prevent aberrant proliferation. Specifically, Bim is
overexpressed in
Myc transgenic mice, determining extensive apoptosis of normal cells. However,
the onset of
tumors in these mice coincides with the loss of one Bim allele that becomes
insufficient. Still,
Bim remains definitely overexpressed in tumors compared to healthy tissues
that are not
subject to oncogenic stress (Egle et al., 2004). Therefore, it is hard to
define a threshold
below which Bim insufficiency occurs and alternative strategies are needed to
define the
importance of miR-25 upregulation in vivo.
[000169] Several mechanisms have been described leading to Bim downregulation
in cancer,
from transcriptional regulation to protein degradation (Yano et al., 2006; Tan
et al., 2005).
While all of these mechanisms clearly contribute to Bim silencing, we propose
miR-25
interference as a novel mechanism of Bim post-transcriptional regulation in
gastric cancer.
[000170] It has been extensively debated whether miRNAs are just fine-tuning
molecules or
they act as key gene switches. Recent studies suggest that both hypotheses are
probably true,
depending on the specific biological context. From this perspective, the
therapeutic potential
of miRNAs in cancer may be strictly associated with the occurrence of specific
miRNA-
dependent functional alterations. Knowing the mechanisms of action of tumor-
related
miRNAs is useful in establishing the molecular diagnosis of miRNA-dependent
tumors,
allowing the rational selection of those patients eventually responding to
miRNA-based
therapies.
[000171] Experimental procedures
[000172] Cell culture and treatments:
[000173] All cell lines were obtained by ATCC and cultured in RPMI 1640 medium
supplemented with 10% fetal bovine serum, penicillin and streptomycin. Cells
were
transfected with Lipofectamine 2000 (Invitrogen) using 100 nM microRNA
precursors
(Ambion), 100 nM si-p21 (Santa Cruz), 100 nM si-Bim (Cell Signalling) or 100
nM LNA
microRNA antisense oligonucleotides (Exiqon). Protein lysates and total RNA
were collected
at the time indicated. MiRNA processing and expression were verified by
Northern Blot and
stem-loop qRT-PCR. We confirmed transfection efficiency (> 95%) using BLOCK-IT
Fluorescent Oligo (Invitrogen) for all the cell lines.

27


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000174] For synchronization experiments, AGS cells were grown in 10% FBS RPMI
1640
containing 0.03 Pg/ml nocodazole for 12 hours and subsequently released in
fresh medium.
Progression through the cell cycle was followed by FACS analysis until 8
hours, after which
cells rapidly lost synchronization.
[000175] For TGFE experiments, 2x106 Snu-16 cells were transfected in 6-well
plates in a
1:1 mixture of Optimem (GIBCO) and RPMI 1640 10% FBS (Sigma) using 5 ul
Lipofectamine 2000 and 100 nM miRNA precursors (Ambion) or LNA antisense
oligonucleotides (Exiqon). After 12 hours, medium was replaced with RPMI 1640
10% FBS
containing 1 ng/ml human recombinant TGFE1 (Sigma). Number of viable cells was
assayed
using WST tetrazolium salt (CCK-8, Dojindo) as per the manufacturer
instructions. All the
experiments were performed in triplicate. Results were expressed as mean SD.
[000176] qRT- PCR:
[000177] Mature miRNAs and other mRNAs were assayed using the single tube
TaqMan
MicroRNA Assays and the Gene Expression Assays, respectively, in accordance
with
manufacturer's instructions (Applied Biosystems, Foster City, CA). All RT
reactions,
including no-template controls and RT minus controls, were run in a GeneAmp
PCR 9700
Thermocycler (Applied Biosystems). RNA concentrations were determined with a
NanoDrop
(NanoDrop Technologies, Inc.). Samples were normalized to RNU49 or CAPN2
(Applied
Biosystems), as indicated. Gene expression levels were quantified using the
ABI Prism
7900HT Sequence detection system (Applied Biosystems). Comparative real-time
PCR was
performed in triplicate, including no-template controls. Relative expression
was calculated
using the comparative Ct method.
[000178] Luciferase Assays
[000179] MKN-74 gastric cancer cells were cotransfected in six-well plates
with 1 ug of
pGL3 firefly luciferase reporter vector (see supplementary Experimental
Procedures), 0.1 ug
of the phRLSV40 control vector (Promega) and 100 nM microRNA precursors
(Ambion)
using Lipofectamine 2000 (Invitrogen). Firefly and Renilla luciferase
activities were
measured consecutively by using the Dual Luciferase Assay (Promega) 24 h after
transfection.
Each reporter plasmid was transfected at least twice (on different days) and
each sample was
assayed in triplicate.
[000180] Flow Cytometry
[000181] For cell cycle analysis, 2x106 cells were fixed in cold methanol,
RNAse-treated,
and stained with propidium iodide (Sigma). Cells were analyzed for DNA content
by EPICS-
28


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
XL scan (Beckman Coulter) by using doublet discrimination gating. All analyses
were
performed in triplicate and 20,000 gated events/sample were counted. For
apoptosis analysis,
cells were washed in cold PBS, incubated with AnnexinV-FITC (BD Biopharmingen)
and PI
(Sigma) for 15 minutes in the dark and analyzed within 1 hour.
[000182] Statistical analysis
[000183] Results of experiments are expressed as mean +/- SD. Student's
unpaired t test
was used to compare values of test and control samples. P < 0.05 indicated
significant
difference.
[000184] EXAMPLE II
[000185] Tissue samples:
[000186] Primary gastric tumor samples were obtained by the Department of
Histopathology
(Sant'Andrea Hospital, University of Rome "La Sapienza", Italy). All of the
samples had
patient's informed consent and were histologically confirmed. Protocol for
tissue
procurement was approved by the Sant'Andrea Hospital Bioethical Committee.
Each tumor
was paired with a non-tumor gastric mucosa control from the same patient.
[000187] Microarrays:
[000188] Microarray analysis was performed as described (Liu et al., 2004).
Briefly, 5 ug of
total RNA was used for hybridization on 2nd generation miRNA microarray chips
(V2).
These chips contain gene-specific 40-mer oligonucleotide probes for 250 human
miRNAs,
spotted by contacting technologies and covalently attached to a polymeric
matrix. The
microarrays were hybridized in 6X SSPE (0.9 M NaCI_60 mM NaH2PO4_H20_8 mM
EDTA, pH 7.4), 30% formamide at 25 C for 18 h, washed in 0.75X TNT
(Tris/HC1/NaC1/Tween 20) at 37 C for 40 min, and processed by using a method
of direct
detection of the biotin-containing transcripts by streptavidin-Alexa Fluor 647
conjugate.
Processed slides were scanned by using a microarray scanner, with the laser
set to 635 nm, at
fixed PMT setting, and a scan resolution of 10 mm. Array data were normalized
using Global
Median, Lowess or Quantile methods, obtaining similar results. Data published
in this study
were derived from Quantile normalization. Differentially expressed miRNAs were
identified
by using the t test procedure within significance analysis of microarrays
(SAM).
[000189] Western Blots:
[000190] Antibodies for immunoblotting were as follows: E2F1 (Santa Cruz,
mouse
monoclonal, 1:500), AIB-1 (Cell Signalling, mouse monoclonal, 1:1000), p21
(Cell
Signalling, mouse monoclonal, 1:1000), p27 (Santa Cruz, mouse monoclonal
1:500), CDK2

29


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
(Cell Signalling, mouse monoclonal, 1:1000), CDK4 (Cell Signalling, mouse
monoclonal,
1:1000), cyclin D1 (Cell Signalling, mouse monoclonal, 1:1000), cyclin E
(Santa Cruz, rabbit
polyclonal, 1:500) p15 (Cell Signalling, rabbit polyclonal, 1:1000), Bim (Cell
Signalling,
rabbit polyclonal 1:1000), Vinculin (Santa Cruz, mouse monoclonal, 1:500),
GAPDH
(Calbiochem, mouse monoclonal, 1:3000). Bands were quantified using GelDoc
software
(Biorad).
[000191] Adenoviral and Lentiviral infections:
[000192] Adeno-E2F1 was kindly provided by G. Leone and infection was
performed as
described (Leone et al., 1998). MiR-106b, miR-93, miR-25 and miR-106b-25
precursor
cDNAs were PCR-amplified from 293T/17 cells genomic DNA and cloned under a CMV
promoter into a variant third-generation lentiviral vector, pRRL-CMV-PGK-GFP-
WPRE,
called Tween, to simultaneously transduce both the reporter GFP and the miRNA.
Lentiviral
supernatants preparation and infection were performed as described (Bonci et
al., 2003).
Lentiviral transduction produced a 710 fold-change in miRNA expression, as
determined by
qRT-PCR. Transduction efficiency >90% was verified by fluorescent microscopy.
[000193] qRT-PCR (miRNA precursors):
[000194] For microRNA precursor qRT-PCR, total RNA isolated with TRIzol
reagent
(Invitrogen) was processed after DNase treatment (Ambion) directly to cDNA by
reverse
transcription using ThermoScript kit (Invitrogen). Target sequences were
amplified by qPCR
using Power Syb-Green PCR Master Mix (Applied Biosystems). Samples were
normalized to
U6. Primer sequences are available upon request.
[000195] Sensor plasmids:
[000196] E2F1, p21 and Bim 3'UTRs containing predicted miRNA binding sites
were
amplified by PCR from genomic DNA (293T/17 cells) and inserted into the pGL3
control
vector (Promega) by using the Xba-I site immediately downstream from the stop
codon of
firefly luciferase. Deletions of the first 3 nucleotides in the miRNA seed-
region
complementary sites were inserted in mutant constructs using QuikChange-site-
directed
mutagenesis kit (Stratagene), according to the manufacturer's protocol. Primer
sequences are
available upon request.
[000197] EXAMPLES of USES and DEFINITIONS THEREOF
[000198] The practice of the present invention will employ, unless otherwise
indicated,
conventional methods of pharmacology, chemistry, biochemistry, recombinant DNA
techniques and immunology, within the skill of the art. Such techniques are
explained fully in



CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
the literature. See, e.g., Handbook of Experimental Immunology, Vols. I-IV (D.
M. Weir and
C. C. Blackwell eds., Blackwell Scientific Publications); A. L. Lehninger,
Biochemistry
(Worth Publishers, Inc., current addition); Sambrook, et al., Molecular
Cloning: A Laboratory
Manual (2nd Edition, 1989); Methods In Enzymology (S. Colowick and N. Kaplan
eds.,
Academic Press, Inc.).
[000199] As such, the definitions herein are provided for further explanation
and are not to
be construed as limiting.
[000200] The articles "a" and "an" are used herein to refer to one or to more
than one (i.e., to
at least one) of the grammatical object of the article. By way of example, "an
element" means
one element or more than one element.
[000201] A "marker" and "biomarker" is a gene and/or protein and/or functional
variants
thereof whose altered level of expression in a tissue or cell from its
expression level in normal
or healthy tissue or cell is associated with a disorder and/or disease state.
[000202] The "normal" level of expression of a marker is the level of
expression of the
marker in cells of a human subject or patient not afflicted with a disorder
and/or disease state.
[000203] An "over-expression" or "significantly higher level of expression" of
a marker
refers to an expression level in a test sample that is greater than the
standard error of the assay
employed to assess expression, and in certain embodiments, at least twice, and
in other
embodiments, three, four, five or ten times the expression level of the marker
in a control
sample (e.g., sample from a healthy subject not having the marker associated
disorder and/or
disease state) and in certain embodiments, the average expression level of the
marker in
several control samples.
[000204] A "significantly lower level of expression" of a marker refers to an
expression
level in a test sample that is at least twice, and in certain embodiments,
three, four, five or ten
times lower than the expression level of the marker in a control sample (e.g.,
sample from a
healthy subject not having the marker associated disorder and/or disease
state) and in certain
embodiments, the average expression level of the marker in several control
samples.
[000205] A kit is any manufacture (e.g. a package or container) comprising at
least one
reagent, e.g., a probe, for specifically detecting the expression of a marker.
The kit may be
promoted, distributed or sold as a unit for performing the methods of the
present invention.
[000206] "Proteins" encompass marker proteins and their fragments; variant
marker proteins
and their fragments; peptides and polypeptides comprising an at least 15 amino
acid segment
31


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
of a marker or variant marker protein; and fusion proteins comprising a marker
or variant
marker protein, or an at least 15 amino acid segment of a marker or variant
marker protein.
[000207] The compositions, kits and methods described herein have the
following non-
limiting uses, among others:

1) assessing whether a subject is afflicted with a disorder and/or disease
state;
2) assessing the stage of a disorder and/or disease state in a subject;

3) assessing the grade of a disorder and/or disease state in a subject;
4) assessing the nature of a disorder and/or disease state in a subject;

5) assessing the potential to develop a disorder and/or disease state in a
subject;

6) assessing the histological type of cells associated with a disorder and/or
disease
state in a subject;

7) making antibodies, antibody fragments or antibody derivatives that are
useful
for treating a disorder and/or disease state in a subject;

8) assessing the presence of a disorder and/or disease state in a subject's
cells;
9) assessing the efficacy of one or more test compounds for inhibiting a
disorder
and/or disease state in a subject; 1

10) assessing the efficacy of a therapy for inhibiting a disorder and/or
disease state
in a subject;

11) monitoring the progression of a disorder and/or disease state in a
subject;

12) selecting a composition or therapy for inhibiting a disorder and/or
disease state
in a subject;

13) treating a subject afflicted with a disorder and/or disease state;
14) inhibiting a disorder and/or disease state in a subject;

15) assessing the harmful potential of a test compound; and

16) preventing the onset of a disorder and/or disease state in a subject at
risk
therefor.
[000208] Screening Methods

32


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000209] Animal models can be created to enable screening of therapeutic
agents useful for
treating or preventing a disorder and/or disease state in a subject.
Accordingly, the methods
are useful for identifying therapeutic agents for treating or preventing a
disorder and/or
disease state in a subject. The methods comprise administering a candidate
agent to an animal
model made by the methods described herein, assessing at least one response in
the animal
model as compared to a control animal model to which the candidate agent has
not been
administered. If at least one response is reduced in symptoms or delayed in
onset, the
candidate agent is an agent for treating or preventing the disease.
[000210] The candidate agents may be pharmacologic agents already known in the
art or
may be agents previously unknown to have any pharmacological activity. The
agents may be
naturally arising or designed in the laboratory. They may be isolated from
microorganisms,
animals or plants, or may be produced recombinantly, or synthesized by any
suitable chemical
method. They may be small molecules, nucleic acids, proteins, peptides or
peptidomimetics.
In certain embodiments, candidate agents are small organic compounds having a
molecular
weight of more than 50 and less than about 2,500 daltons. Candidate agents
comprise
functional groups necessary for structural interaction with proteins.
Candidate agents are also
found among biomolecules including, but not limited to: peptides, saccharides,
fatty acids,
steroids, purines, pyrimidines, derivatives, structural analogs or
combinations thereof.
[000211] Candidate agents are obtained from a wide variety of sources
including libraries of
synthetic or natural compounds. There are, for example, numerous means
available for
random and directed synthesis of a wide variety of organic compounds and
biomolecules,
including expression of randomized oligonucleotides and oligopeptides.
Alternatively,
libraries of natural compounds in the form of bacterial, fungal, plant and
animal extracts are
available or readily produced. Additionally, natural or synthetically produced
libraries and
compounds are readily modified through conventional chemical, physical and
biochemical
means, and may be used to produce combinatorial libraries. In certain
embodiments, the
candidate agents can be obtained using any of the numerous approaches in
combinatorial
library methods art, including, by non-limiting example: biological libraries;
spatially
addressable parallel solid phase or solution phase libraries; synthetic
library methods requiring
deconvolution; the "one-bead one-compound" library method; and synthetic
library methods
using affinity chromatography selection.

33


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000212] In certain further embodiments, certain pharmacological agents may be
subjected
to directed or random chemical modifications, such as acylation, alkylation,
esterification,
amidification, etc. to produce structural analogs.
[000213] The same methods for identifying therapeutic agents for treating a
disorder and/or
disease state in a subject can also be used to validate lead compounds/agents
generated from
in vitro studies.
[000214] The candidate agent may be an agent that up- or down-regulates one or
more a
disorder and/or disease state in a subject response pathway. In certain
embodiments, the
candidate agent may be an antagonist that affects such pathway.
[000215] Methods for Treating a Disorder and/or Disease State
[000216] There is provided herein methods for treating, inhibiting, relieving
or reversing a
disorder and/or disease state response. In the methods described herein, an
agent that
interferes with a signaling cascade is administered to an individual in need
thereof, such as,
but not limited to, subjects in whom such complications are not yet evident
and those who
already have at least one such response.
[000217] In the former instance, such treatment is useful to prevent the
occurrence of such
response and/or reduce the extent to which they occur. In the latter instance,
such treatment is
useful to reduce the extent to which such response occurs, prevent their
further development
or reverse the response.
[000218] In certain embodiments, the agent that interferes with the response
cascade may be
an antibody specific for such response.
[000219] Expression of Biomarker(s)
[000220] Expression of a marker can be inhibited in a number of ways,
including, by way of
a non-limiting example, an antisense oligonucleotide can be provided to the
disease cells in
order to inhibit transcription, translation, or both, of the marker(s).
Alternately, a
polynucleotide encoding an antibody, an antibody derivative, or an antibody
fragment which
specifically binds a marker protein, and operably linked with an appropriate
promoter/regulator region, can be provided to the cell in order to generate
intracellular
antibodies which will inhibit the function or activity of the protein. The
expression and/or
function of a marker may also be inhibited by treating the disease cell with
an antibody,
antibody derivative or antibody fragment that specifically binds a marker
protein. Using the
methods described herein, a variety of molecules, particularly including
molecules sufficiently
small that they are able to cross the cell membrane, can be screened in order
to identify

34


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
molecules which inhibit expression of a marker or inhibit the function of a
marker protein.
The compound so identified can be provided to the subject in order to inhibit
disease cells of
the subject.
[000221] Any marker or combination of markers, as well as any certain markers
in
combination with the markers, may be used in the compositions, kits and
methods described
herein. In general, it is desirable to use markers for which the difference
between the level of
expression of the marker in disease cells and the level of expression of the
same marker in
normal colon system cells is as great as possible. Although this difference
can be as small as
the limit of detection of the method for assessing expression of the marker,
it is desirable that
the difference be at least greater than the standard error of the assessment
method, and, in
certain embodiments, a difference of at least 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-,
10-, 15-, 20-, 100-,
500-, 1000-fold or greater than the level of expression of the same marker in
normal tissue.
[000222] It is recognized that certain marker proteins are secreted to the
extracellular space
surrounding the cells. These markers are used in certain embodiments of the
compositions,
kits and methods, owing to the fact that such marker proteins can be detected
in a body fluid
sample, which may be more easily collected from a human subject than a tissue
biopsy
sample. In addition, in vivo techniques for detection of a marker protein
include introducing
into a subject a labeled antibody directed against the protein. For example,
the antibody can
be labeled with a radioactive marker whose presence and location in a subject
can be detected
by standard imaging techniques.
[000223] In order to determine whether any particular marker protein is a
secreted protein,
the marker protein is expressed in, for example, a mammalian cell, such as a
human cell line,
extracellular fluid is collected, and the presence or absence of the protein
in the extracellular
fluid is assessed (e.g. using a labeled antibody which binds specifically with
the protein).
[000224] It will be appreciated that subject samples containing such cells may
be used in the
methods described herein. In these embodiments, the level of expression of the
marker can be
assessed by assessing the amount (e.g., absolute amount or concentration) of
the marker in a
sample. The cell sample can, of course, be subjected to a variety of post-
collection
preparative and storage techniques (e.g., nucleic acid and/or protein
extraction, fixation,
storage, freezing, ultrafiltration, concentration, evaporation,
centrifugation, etc.) prior to
assessing the amount of the marker in the sample.
[000225] It will also be appreciated that the markers may be shed from the
cells into, for
example, the respiratory system, digestive system, the blood stream and/or
interstitial spaces.


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
The shed markers can be tested, for example, by examining the sputum, BAL,
serum, plasma,
urine, stool, etc.
[000226] The compositions, kits and methods can be used to detect expression
of marker
proteins having at least one portion which is displayed on the surface of
cells which express it.
For example, immunological methods may be used to detect such proteins on
whole cells, or
computer-based sequence analysis methods may be used to predict the presence
of at least one
extracellular domain (i.e., including both secreted proteins and proteins
having at least one
cell-surface domain). Expression of a marker protein having at least one
portion which is
displayed on the surface of a cell which expresses it may be detected without
necessarily
lysing the cell (e.g., using a labeled antibody which binds specifically with
a cell-surface
domain of the protein).
[000227] Expression of a marker may be assessed by any of a wide variety of
methods for
detecting expression of a transcribed nucleic acid or protein. Non-limiting
examples of such
methods include immunological methods for detection of secreted, cell-surface,
cytoplasmic
or nuclear proteins, protein purification methods, protein function or
activity assays, nucleic
acid hybridization methods, nucleic acid reverse transcription methods and
nucleic acid
amplification methods.
[000228] In a particular embodiment, expression of a marker is assessed using
an antibody
(e.g., a radio-labeled, chromophore-labeled, fluorophore-labeled or enzyme-
labeled antibody),
an antibody derivative (e.g., an antibody conjugated with a substrate or with
the protein or
ligand of a protein-ligand pair), or an antibody fragment (e.g., a single-
chain antibody, an
isolated antibody hypervariable domain, etc.) which binds specifically with a
marker protein
or fragment thereof, including a marker protein which has undergone all or a
portion of its
normal post-translational modification.
[000229] In another particular embodiment, expression of a marker is assessed
by preparing
mRNA/cDNA (i.e., a transcribed polynucleotide) from cells in a subject sample,
and by
hybridizing the mRNA/cDNA with a reference polynucleotide which is a
complement of a
marker nucleic acid, or a fragment thereof. cDNA can, optionally, be amplified
using any of a
variety of polymerase chain reaction methods prior to hybridization with the
reference
polynucleotide; preferably, it is not amplified. Expression of one or more
markers can
likewise be detected using quantitative PCR to assess the level of expression
of the marker(s).
Alternatively, any of the many methods of detecting mutations or variants
(e.g., single

36


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
nucleotide polymorphisms, deletions, etc.) of a marker may be used to detect
occurrence of a
marker in a subject.
[000230] In a related embodiment, a mixture of transcribed polynucleotides
obtained from
the sample is contacted with a substrate having fixed thereto a polynucleotide
complementary
to or homologous with at least a portion (e.g., at least 7, 10, 15, 20, 25,
30, 40, 50, 100, 500, or
more nucleotide residues) of a marker nucleic acid. If polynucleotides
complementary to or
homologous with are differentially detectable on the substrate (e.g.,
detectable using different
chromophores or fluorophores, or fixed to different selected positions), then
the levels of
expression of a plurality of markers can be assessed simultaneously using a
single substrate
(e.g., a "gene chip" microarray of polynucleotides fixed at selected
positions). When a
method of assessing marker expression is used which involves hybridization of
one nucleic
acid with another, it is desired that the hybridization be performed under
stringent
hybridization conditions.
[000231] In certain embodiments, the biomarker assays can be performed using
mass
spectrometry or surface plasmon resonance. In various embodiment, the method
of
identifying an agent active against a disorder and/or disease state in a
subject can include one
or more of:

a) providing a sample of cells containing one or more markers or derivative
thereof;

b) preparing an extract from such cells;

c) mixing the extract with a labeled nucleic acid probe containing a marker
binding
site; and,

d) determining the formation of a complex between the marker and the nucleic
acid
probe in the presence or absence of the test agent. The determining step can
include
subjecting said extract/nucleic acid probe mixture to an electrophoretic
mobility shift
assay.
[000232] In certain embodiments, the determining step comprises an assay
selected from an
enzyme linked immunoabsorption assay (ELISA), fluorescence based assays and
ultra high
throughput assays, for example surface plasmon resonance (SPR) or fluorescence
correlation
spectroscopy (FCS) assays. In such embodiments, the SPR sensor is useful for
direct real-
time observation of biomolecular interactions since SPR is sensitive to minute
refractive index

37


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
changes at a metal-dielectric surface. SPR is a surface technique that is
sensitive to changes
of 105 to 10.6 refractive index (RI) units within approximately 200 nm of the
SPR
sensor/sample interface. Thus, SPR spectroscopy is useful for monitoring the
growth of thin
organic films deposited on the sensing layer.
[000233] Because the compositions, kits, and methods rely on detection of a
difference in
expression levels of one or more markers, it is desired that the level of
expression of the
marker is significantly greater than the minimum detection limit of the method
used to assess
expression in at least one of normal cells and colon cancer-affected cells.
[000234] It is understood that by routine screening of additional subject
samples using one
or more of the markers, it will be realized that certain of the markers are
over-expressed in
cells of various types, including a specific disorders and/or disease state in
a subject.
[000235] In addition, as a greater number of subject samples are assessed for
expression of
the markers and the outcomes of the individual subjects from whom the samples
were
obtained are correlated, it will also be confirmed that altered expression of
certain of the
markers are strongly correlated with a disorder and/or disease state in a
subject and that
altered expression of other markers are strongly correlated with other
diseases. The
compositions, kits, and methods are thus useful for characterizing one or more
of the stage,
grade, histological type, and nature of a disorder and/or disease state in a
subject.
[000236] When the compositions, kits, and methods are used for characterizing
one or more
of the stage, grade, histological type, and nature of a disorder and/or
disease state in a subject,
it is desired that the marker or panel of markers is selected such that a
positive result is
obtained in at least about 20%, and in certain embodiments, at least about
40%, 60%, or 80%,
and in substantially all subjects afflicted with a disorder and/or disease
state of the
corresponding stage, grade, histological type, or nature. The marker or panel
of markers
invention can be selected such that a positive predictive value of greater
than about 10% is
obtained for the general population (in a non-limiting example, coupled with
an assay
specificity greater than 80%).
[000237] When a plurality of markers are used in the compositions, kits, and
methods, the
level of expression of each marker in a subject sample can be compared with
the normal level
of expression of each of the plurality of markers in non- disorder and/or non-
disease samples
of the same type, either in a single reaction mixture (i.e. using reagents,
such as different
fluorescent probes, for each marker) or in individual reaction mixtures
corresponding to one
or more of the markers. In one embodiment, a significantly increased level of
expression of

38


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
more than one of the plurality of markers in the sample, relative to the
corresponding normal
levels, is an indication that the subject is afflicted with a disorder and/or
disease state. When a
plurality of markers is used, 2, 3, 4, 5, 8, 10, 12, 15, 20, 30, or 50 or more
individual markers
can be used; in certain embodiments, the use of fewer markers may be desired.
[000238] In order to maximize the sensitivity of the compositions, kits, and
methods (i.e. by
interference attributable to cells of system origin in a subject sample), it
is desirable that the
marker used therein be a marker which has a restricted tissue distribution,
e.g., normally not
expressed in a non-system tissue.
[000239] It is recognized that the compositions, kits, and methods will be of
particular utility
to subjects having an enhanced risk of developing a disorder and/or disease
state in a subject
and their medical advisors. Subjects recognized as having an enhanced risk of
developing a
disorder and/or disease include, for example, subjects having a familial
history of such
disorder or disease.
[000240] The level of expression of a marker in normal human system tissue can
be assessed
in a variety of ways. In one embodiment, this normal level of expression is
assessed by
assessing the level of expression of the marker in a portion of system cells
which appear to be
normal and by comparing this normal level of expression with the level of
expression in a
portion of the system cells which is suspected of being abnormal. Alternately,
and
particularly as further information becomes available as a result of routine
performance of the
methods described herein, population-average values for normal expression of
the markers
may be used. In other embodiments, the 'normal' level of expression of a
marker may be
determined by assessing expression of the marker in a subject sample obtained
from a non-
afflicted subject, from a subject sample obtained from a subject before the
suspected onset of
a disorder and/or disease state in the subject, from archived subject samples,
and the like.
[000241] There is also provided herein compositions, kits, and methods for
assessing the
presence of disorder and/or disease state cells in a sample (e.g. an archived
tissue sample or a
sample obtained from a subject). These compositions, kits, and methods are
substantially the
same as those described above, except that, where necessary, the compositions,
kits, and
methods are adapted for use with samples other than subject samples. For
example, when the
sample to be used is a parafinized, archived human tissue sample, it can be
necessary to adjust
the ratio of compounds in the compositions, in the kits, or the methods used
to assess levels of
marker expression in the sample.
[000242] Kits and Reagents

39


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000243] The kits are useful for assessing the presence of disease cells (e.g.
in a sample such
as a subject sample). The kit comprises a plurality of reagents, each of which
is capable of
binding specifically with a marker nucleic acid or protein. Suitable reagents
for binding with
a marker protein include antibodies, antibody derivatives, antibody fragments,
and the like.
Suitable reagents for binding with a marker nucleic acid (e.g. a genomic DNA,
an MRNA, a
spliced MRNA, a cDNA, or the like) include complementary nucleic acids. For
example, the
nucleic acid reagents may include oligonucleotides (labeled or non-labeled)
fixed to a
substrate, labeled oligonucleotides not bound with a substrate, pairs of PCR
primers,
molecular beacon probes, and the like.
[000244] The kits may optionally comprise additional components useful for
performing the
methods described herein. By way of example, the kit may comprise fluids (e.g.
SSC buffer)
suitable for annealing complementary nucleic acids or for binding an antibody
with a protein
with which it specifically binds, one or more sample compartments, an
instructional material
which describes performance of the method, a sample of normal colon system
cells, a sample
of colon cancer-related disease cells, and the like.
[000245] Methods of Producing Antibodies
[000246] There is also provided herein a method of making an isolated
hybridoma which
produces an antibody useful for assessing whether a subject is afflicted with
a disorder and/or
disease state. In this method, a protein or peptide comprising the entirety or
a segment of a
marker protein is synthesized or isolated (e.g. by purification from a cell in
which it is
expressed or by transcription and translation of a nucleic acid encoding the
protein or peptide
in vivo or in vitro). A vertebrate, for example, a mammal such as a mouse,
rat, rabbit, or
sheep, is immunized using the protein or peptide. The vertebrate may
optionally (and
preferably) be immunized at least one additional time with the protein or
peptide, so that the
vertebrate exhibits a robust immune response to the protein or peptide.
Splenocytes are
isolated from the immunized vertebrate and fused with an immortalized cell
line to form
hybridomas, using any of a variety of methods. Hybridomas formed in this
manner are then
screened using standard methods to identify one or more hybridomas which
produce an
antibody which specifically binds with the marker protein or a fragment
thereof. There is also
provided herein hybridomas made by this method and antibodies made using such
hybridomas.
[000247] Methods of Assessing Efficacy



CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000248] There is also provided herein a method of assessing the efficacy of a
test
compound for inhibiting disease cells. As described above, differences in the
level of
expression of the markers correlate with the abnormal state of the subject's
cells. Although it
is recognized that changes in the levels of expression of certain of the
markers likely result
from the abnormal state of such cells, it is likewise recognized that changes
in the levels of
expression of other of the markers induce, maintain, and promote the abnormal
state of those
cells. Thus, compounds which inhibit a disorder and/or disease state in a
subject will cause
the level of expression of one or more of the markers to change to a level
nearer the normal
level of expression for that marker (i.e. the level of expression for the
marker in normal cells).
[000249] This method thus comprises comparing expression of a marker in a
first cell
sample and maintained in the presence of the test compound and expression of
the marker in a
second colon cell sample and maintained in the absence of the test compound. A
significantly
reduced expression of a marker in the presence of the test compound is an
indication that the
test compound inhibits a related disease. The cell samples may, for example,
be aliquots of a
single sample of normal cells obtained from a subject, pooled samples of
normal cells
obtained from a subject, cells of a normal cell line, aliquots of a single
sample of related
disease cells obtained from a subject, pooled samples of related disease cells
obtained from a
subject, cells of a related disease cell line, or the like.
[000250] In one embodiment, the samples are cancer-related disease cells
obtained from a
subject and a plurality of compounds believed to be effective for inhibiting
various cancer-
related diseases are tested in order to identify the compound which is likely
to best inhibit the
cancer-related disease in the subject.
[000251] This method may likewise be used to assess the efficacy of a therapy
for inhibiting
a related disease in a subject. In this method, the level of expression of one
or more markers
in a pair of samples (one subjected to the therapy, the other not subjected to
the therapy) is
assessed. As with the method of assessing the efficacy of test compounds, if
the therapy
induces a significantly lower level of expression of a marker then the therapy
is efficacious for
inhibiting a cancer-related disease. As above, if samples from a selected
subject are used in
this method, then alternative therapies can be assessed in vitro in order to
select a therapy
most likely to be efficacious for inhibiting a cancer-related disease in the
subject.
[000252] As described herein, the abnormal state of human cells is correlated
with changes
in the levels of expression of the markers. There is also provided a method
for assessing the
harmful potential of a test compound. This method comprises maintaining
separate aliquots
41


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
of human cells in the presence and absence of the test compound. Expression of
a marker in
each of the aliquots is compared. A significantly higher level of expression
of a marker in the
aliquot maintained in the presence of the test compound (relative to the
aliquot maintained in
the absence of the test compound) is an indication that the test compound
possesses a harmful
potential. The relative harmful potential of various test compounds can be
assessed by
comparing the degree of enhancement or inhibition of the level of expression
of the relevant
markers, by comparing the number of markers for which the level of expression
is enhanced
or inhibited, or by comparing both. Various aspects are described in further
detail in the
following subsections.
[000253] Isolated Proteins and Antibodies
[000254] One aspect pertains to isolated marker proteins and biologically
active portions
thereof, as well as polypeptide fragments suitable for use as immunogens to
raise antibodies
directed against a marker protein or a fragment thereof. In one embodiment,
the native marker
protein can be isolated from cells or tissue sources by an appropriate
purification scheme
using standard protein purification techniques. In another embodiment, a
protein or peptide
comprising the whole or a segment of the marker protein is produced by
recombinant DNA
techniques. Alternative to recombinant expression, such protein or peptide can
be synthesized
chemically using standard peptide synthesis techniques.
[000255] An "isolated" or "purified" protein or biologically active portion
thereof is
substantially free of cellular material or other contaminating proteins from
the cell or tissue
source from which the protein is derived, or substantially free of chemical
precursors or other
chemicals when chemically synthesized. The language "substantially free of
cellular material"
includes preparations of protein in which the protein is separated from
cellular components of
the cells from which it is isolated or recombinantly produced. Thus, protein
that is
substantially free of cellular material includes preparations of protein
having less than about
30%, 20%, 10%, or 5% (by dry weight) of heterologous protein (also referred to
herein as a
"contaminating protein").
[000256] When the protein or biologically active portion thereof is
recombinantly produced,
it is also preferably substantially free of culture medium, i.e., culture
medium represents less
than about 20%, 10%, or 5% of the volume of the protein preparation. When the
protein is
produced by chemical synthesis, it is preferably substantially free of
chemical precursors or
other chemicals, i.e., it is separated from chemical precursors or other
chemicals which are
involved in the synthesis of the protein. Accordingly such preparations of the
protein have

42


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
less than about 30%, 20%, 10%, 5% (by dry weight) of chemical precursors or
compounds
other than the polypeptide of interest.
[000257] Biologically active portions of a marker protein include polypeptides
comprising
amino acid sequences sufficiently identical to or derived from the amino acid
sequence of the
marker protein, which include fewer amino acids than the full length protein,
and exhibit at
least one activity of the corresponding full-length protein. Typically,
biologically active
portions comprise a domain or motif with at least one activity of the
corresponding full-length
protein. A biologically active portion of a marker protein can be a
polypeptide which is, for
example, 10, 25, 50, 100 or more amino acids in length. Moreover, other
biologically active
portions, in which other regions of the marker protein are deleted, can be
prepared by
recombinant techniques and evaluated for one or more of the functional
activities of the native
form of the marker protein. In certain embodiments, useful proteins are
substantially identical
(e.g., at least about 40%, and in certain embodiments, 50%, 60%, 70%, 80%,
90%, 95%, or
99%) to one of these sequences and retain the functional activity of the
corresponding
naturally-occurring marker protein yet differ in amino acid sequence due to
natural allelic
variation or mutagenesis.
[000258] In addition, libraries of segments of a marker protein can be used to
generate a
variegated population of polypeptides for screening and subsequent selection
of variant
marker proteins or segments thereof.
[000259] Predictive Medicine
[000260] There is also provided herein uses of the animal models and markers
in the field of
predictive medicine in which diagnostic assays, prognostic assays,
pharmacogenomics, and
monitoring clinical trials are used for prognostic (predictive) purposes to
thereby treat an
individual prophylactically. Accordingly, there is also provided herein
diagnostic assays for
determining the level of expression of one or more marker proteins or nucleic
acids, in order
to determine whether an individual is at risk of developing a particular
disorder and/or disease.
Such assays can be used for prognostic or predictive purposes to thereby
prophylactically treat
an individual prior to the onset of the disorder and/or disease.
[000261] In another aspect, the methods are useful for at least periodic
screening of the same
individual to see if that individual has been exposed to chemicals or toxins
that change his/her
expression patterns.
[000262] Yet another aspect pertains to monitoring the influence of agents
(e.g., drugs or
other compounds administered either to inhibit a disorder and/or disease or to
treat or prevent
43


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
any other disorder (e.g., in order to understand any system effects that such
treatment may
have) on the expression or activity of a marker in clinical trials.
[000263] Pharmaceutical Compositions
[000264] The compounds may be in a formulation for administration topically,
locally or
systemically in a suitable pharmaceutical carrier. Remington's Pharmaceutical
Sciences, 15th
Edition by E. W. Martin (Mark Publishing Company, 1975), discloses typical
carriers and
methods of preparation. The compound may also be encapsulated in suitable
biocompatible
microcapsules, microparticles or micro spheres formed of biodegradable or non-
biodegradable
polymers or proteins or liposomes for targeting to cells. Such systems are
well known to
those skilled in the art and may be optimized for use with the appropriate
nucleic acid.
[000265] Various methods for nucleic acid delivery are described, for example
in Sambrook
et al., 1989, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor
Laboratory, New
York; and Ausubel et al., 1994, Current Protocols in Molecular Biology, John
Wiley & Sons,
New York. Such nucleic acid delivery systems comprise the desired nucleic
acid, by way of
example and not by limitation, in either "naked" form as a "naked" nucleic
acid, or formulated
in a vehicle suitable for delivery, such as in a complex with a cationic
molecule or a liposome
forming lipid, or as a component of a vector, or a component of a
pharmaceutical
composition. The nucleic acid delivery system can be provided to the cell
either directly, such
as by contacting it with the cell, or indirectly, such as through the action
of any biological
process.
[000266] Formulations for topical administration may include ointments,
lotions, creams,
gels, drops, suppositories, sprays, liquids and powders. Conventional
pharmaceutical carriers,
aqueous, powder or oily bases, or thickeners can be used as desired.
[000267] Formulations suitable for parenteral administration, such as, for
example, by
intraarticular (in the joints), intravenous, intramuscular, intradermal,
intraperitoneal, and
subcutaneous routes, include aqueous and non-aqueous, isotonic sterile
injection solutions,
which can contain antioxidants, buffers, bacteriostats, and solutes that
render the formulation
isotonic with the blood of the intended recipient, and aqueous and non-aqueous
sterile
suspensions, solutions or emulsions that can include suspending agents,
solubilizers,
thickening agents, dispersing agents, stabilizers, and preservatives.
Formulations for injection
may be presented in unit dosage form, e.g., in ampules or in multi-dose
containers, with an
added preservative. Those of skill in the art can readily determine the
various parameters for

44


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
preparing and formulating the compositions without resort to undue
experimentation. The
compound can be used alone or in combination with other suitable components.
[000268] In general, methods of administering compounds, including nucleic
acids, are well
known in the art. In particular, the routes of administration already in use
for nucleic acid
therapeutics, along with formulations in current use, provide preferred routes
of administration
and formulation for the nucleic acids selected will depend of course, upon
factors such as the
particular formulation, the severity of the state of the subject being
treated, and the dosage
required for therapeutic efficacy. As generally used herein, an "effective
amount" is that
amount which is able to treat one or more symptoms of the disorder, reverse
the progression
of one or more symptoms of the disorder, halt the progression of one or more
symptoms of the
disorder, or prevent the occurrence of one or more symptoms of the disorder in
a subject to
whom the formulation is administered, as compared to a matched subject not
receiving the
compound. The actual effective amounts of compound can vary according to the
specific
compound or combination thereof being utilized, the particular composition
formulated, the
mode of administration, and the age, weight, condition of the individual, and
severity of the
symptoms or condition being treated.
[000269] Any acceptable method known to one of ordinary skill in the art may
be used to
administer a formulation to the subject. The administration may be localized
(i.e., to a
particular region, physiological system, tissue, organ, or cell type) or
systemic, depending on
the condition being treated.
[000270] Pharmacogenomics
[000271] The markers are also useful as pharmacogenomic markers. As used
herein, a
"pharmacogenomic marker" is an objective biochemical marker whose expression
level
correlates with a specific clinical drug response or susceptibility in a
subject. The presence or
quantity of the pharmacogenomic marker expression is related to the predicted
response of the
subject and more particularly the subject's tumor to therapy with a specific
drug or class of
drugs. By assessing the presence or quantity of the expression of one or more
pharmacogenomic markers in a subject, a drug therapy which is most appropriate
for the
subject, or which is predicted to have a greater degree of success, may be
selected.
[000272] Monitoring Clinical Trials
[000273] Monitoring the influence of agents (e.g., drug compounds) on the
level of
expression of a marker can be applied not only in basic drug screening, but
also in clinical


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
trials. For example, the effectiveness of an agent to affect marker expression
can be
monitored in clinical trials of subjects receiving treatment for a colon
cancer-related disease.
[000274] In one non-limiting embodiment, the present invention provides a
method for
monitoring the effectiveness of treatment of a subject with an agent (e.g., an
agonist,
antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or
other drug
candidate) comprising the steps of:

i) obtaining a pre-administration sample from a subject prior to
administration of
the agent;

ii) detecting the level of expression of one or more selected markers in the
pre-
administration sample;

iii) obtaining one or more post-administration samples from the subject;

iv) detecting the level of expression of the marker(s) in the post-
administration
samples;

v) comparing the level of expression of the marker(s) in the pre-
administration
sample with the level of expression of the marker(s) in the post-
administration sample or
samples; and

vi) altering the administration of the agent to the subject accordingly.
[000275] For example, increased expression of the marker gene(s) during the
course of
treatment may indicate ineffective dosage and the desirability of increasing
the dosage.
Conversely, decreased expression of the marker gene(s) may indicate
efficacious treatment
and no need to change dosage.
[000276] Electronic Apparatus Readable Media, Systems, Arrays and Methods of
Using
Same
[000277] As used herein, "electronic apparatus readable media" refers to any
suitable
medium for storing, holding or containing data or information that can be read
and accessed
directly by an electronic apparatus. Such media can include, but are not
limited to: magnetic
storage media, such as floppy discs, hard disc storage medium, and magnetic
tape; optical
storage media such as compact disc; electronic storage media such as RAM, ROM,
EPROM,
EEPROM and the like; and general hard disks and hybrids of these categories
such as
magnetic/optical storage media. The medium is adapted or configured for having
recorded
thereon a marker as described herein.

46


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000278] As used herein, the term "electronic apparatus" is intended to
include any suitable
computing or processing apparatus or other device configured or adapted for
storing data or
information. Examples of electronic apparatus suitable for use with the
present invention
include stand-alone computing apparatus; networks, including a local area
network (LAN), a
wide area network (WAN) Internet, Intranet, and Extranet; electronic
appliances such as
personal digital assistants (PDAs), cellular phone, pager and the like; and
local and distributed
processing systems.
[000279] As used herein, "recorded" refers to a process for storing or
encoding information
on the electronic apparatus readable medium. Those skilled in the art can
readily adopt any
method for recording information on media to generate materials comprising the
markers
described herein.
[000280] A variety of software programs and formats can be used to store the
marker
information of the present invention on the electronic apparatus readable
medium. Any
number of data processor structuring formats (e.g., text file or database) may
be employed in
order to obtain or create a medium having recorded thereon the markers. By
providing the
markers in readable form, one can routinely access the marker sequence
information for a
variety of purposes. For example, one skilled in the art can use the
nucleotide or amino acid
sequences in readable form to compare a target sequence or target structural
motif with the
sequence information stored within the data storage means. Search means are
used to identify
fragments or regions of the sequences which match a particular target sequence
or target
motif.
[000281] Thus, there is also provided herein a medium for holding instructions
for
performing a method for determining whether a subject has a cancer-related
disease or a pre-
disposition to a cancer-related disease, wherein the method comprises the
steps of determining
the presence or absence of a marker and based on the presence or absence of
the marker,
determining whether the subject has a cancer-related disease or a pre-
disposition to a cancer-
related disease and/or recommending a particular treatment for a cancer-
related disease or pre-
cancer-related disease condition.
[000282] There is also provided herein an electronic system and/or in a
network, a method
for determining whether a subject has a cancer-related disease or a pre-
disposition to a cancer-
related disease associated with a marker wherein the method comprises the
steps of
determining the presence or absence of the marker, and based on the presence
or absence of
the marker, determining whether the subject has a particular disorder and/or
disease or a pre-

47


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
disposition to such disorder and/or disease, and/or recommending a particular
treatment for
such disease or disease and/or such pre-cancer-related disease condition. The
method may
further comprise the step of receiving phenotypic information associated with
the subject
and/or acquiring from a network phenotypic information associated with the
subject.
[000283] Also provided herein is a network, a method for determining whether a
subject has
a disorder and/or disease or a pre-disposition to a disorder and/or disease
associated with a
marker, the method comprising the steps of receiving information associated
with the marker,
receiving phenotypic information associated with the subject, acquiring
information from the
network corresponding to the marker and/or disorder and/or disease, and based
on one or more
of the phenotypic information, the marker, and the acquired information,
determining whether
the subject has a disorder and/or disease or a pre-disposition thereto. The
method may further
comprise the step of recommending a particular treatment for the disorder
and/or disease or
pre-disposition thereto.
[000284] There is also provided herein a business method for determining
whether a subject
has a disorder and/or disease or a pre-disposition thereto, the method
comprising the steps of
receiving information associated with the marker, receiving phenotypic
information associated
with the subject, acquiring information from the network corresponding to the
marker and/or a
disorder and/or disease, and based on one or more of the phenotypic
information, the marker,
and the acquired information, determining whether the subject has a disorder
and/or disease or
a pre-disposition thereto. The method may further comprise the step of
recommending a
particular treatment therefor.
[000285] There is also provided herein an array that can be used to assay
expression of one
or more genes in the array. In one embodiment, the array can be used to assay
gene
expression in a tissue to ascertain tissue specificity of genes in the array.
In this manner, up to
about 7000 or more genes can be simultaneously assayed for expression. This
allows a profile
to be developed showing a battery of genes specifically expressed in one or
more tissues.
[000286] In addition to such qualitative determination, there is provided
herein the
quantitation of gene expression. Thus, not only tissue specificity, but also
the level of
expression of a battery of genes in the tissue is ascertainable. Thus, genes
can be grouped on
the basis of their tissue expression per se and level of expression in that
tissue. This is useful,
for example, in ascertaining the relationship of gene expression between or
among tissues.
Thus, one tissue can be perturbed and the effect on gene expression in a
second tissue can be

48


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
determined. In this context, the effect of one cell type on another cell type
in response to a
biological stimulus can be determined.
[000287] Such a determination is useful, for example, to know the effect of
cell-cell
interaction at the level of gene expression. If an agent is administered
therapeutically to treat
one cell type but has an undesirable effect on another cell type, the method
provides an assay
to determine the molecular basis of the undesirable effect and thus provides
the opportunity to
co-administer a counteracting agent or otherwise treat the undesired effect.
Similarly, even
within a single cell type, undesirable biological effects can be determined at
the molecular
level. Thus, the effects of an agent on expression of other than the target
gene can be
ascertained and counteracted.
[000288] In another embodiment, the array can be used to monitor the time
course of
expression of one or more genes in the array. This can occur in various
biological contexts, as
disclosed herein, for example development of a disorder and/or disease,
progression thereof,
and processes, such as cellular transformation associated therewith.
[000289] The array is also useful for ascertaining the effect of the
expression of a gene or the
expression of other genes in the same cell or in different cells. This
provides, for example, for
a selection of alternate molecular targets for therapeutic intervention if the
ultimate or
downstream target cannot be regulated.
[000290] The array is also useful for ascertaining differential expression
patterns of one or
more genes in normal and abnormal cells. This provides a battery of genes that
could serve as
a molecular target for diagnosis or therapeutic intervention.
[000291] Surrogate Markers
[000292] The markers may serve as surrogate markers for one or more disorders
or disease
states or for conditions leading up thereto. As used herein, a "surrogate
marker" is an
objective biochemical marker which correlates with the absence or presence of
a disease or
disorder, or with the progression of a disease or disorder. The presence or
quantity of such
markers is independent of the disease. Therefore, these markers may serve to
indicate
whether a particular course of treatment is effective in lessening a disease
state or disorder.
Surrogate markers are of particular use when the presence or extent of a
disease state or
disorder is difficult to assess through standard methodologies, or when an
assessment of
disease progression is desired before a potentially dangerous clinical
endpoint is reached.
[000293] The markers are also useful as pharmacodynamic markers. As used
herein, a
"pharmacodynamic marker" is an objective biochemical marker which correlates
specifically
49


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
with drug effects. The presence or quantity of a pharmacodynamic marker is not
related to the
disease state or disorder for which the drug is being administered; therefore,
the presence or
quantity of the marker is indicative of the presence or activity of the drug
in a subject. For
example, a pharmacodynamic marker may be indicative of the concentration of
the drug in a
biological tissue, in that the marker is either expressed or transcribed or
not expressed or
transcribed in that tissue in relationship to the level of the drug. In this
fashion, the
distribution or uptake of the drug may be monitored by the pharmacodynamic
marker.
Similarly, the presence or quantity of the pharmacodynamic marker may be
related to the
presence or quantity of the metabolic product of a drug, such that the
presence or quantity of
the marker is indicative of the relative breakdown rate of the drug in vivo.
[000294] Pharmacodynamic markers are of particular use in increasing the
sensitivity of
detection of drug effects, particularly when the drug is administered in low
doses. Since even
a small amount of a drug may be sufficient to activate multiple rounds of
marker transcription
or expression, the amplified marker may be in a quantity which is more readily
detectable than
the drug itself. Also, the marker may be more easily detected due to the
nature of the marker
itself; for example, using the methods described herein, antibodies may be
employed in an
immune-based detection system for a protein marker, or marker-specific
radiolabeled probes
may be used to detect a mRNA marker. Furthermore, the use of a pharmacodynamic
marker
may offer mechanism-based prediction of risk due to drug treatment beyond the
range of
possible direct observations.
[000295] Protocols for Testing
[000296] The method of testing for a disorder and/or disease may comprise, for
example
measuring the expression level of each marker gene in a biological sample from
a subject over
time and comparing the level with that of the marker gene in a control
biological sample.
[000297] When the marker gene is one of the genes described herein and the
expression
level is differentially expressed (for examples, higher or lower than that in
the control), the
subject is judged to be affected with a disorder and/or disease. When the
expression level of
the marker gene falls within the permissible range, the subject is unlikely to
be affected
therewith.
[000298] The standard value for the control may be pre-determined by measuring
the
expression level of the marker gene in the control, in order to compare the
expression levels.
For example, the standard value can be determined based on the expression
level of the above-
mentioned marker gene in the control. For example, in certain embodiments, the
permissible



CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
range is taken as 2S.D. based on the standard value. Once the standard value
is determined,
the testing method may be performed by measuring only the expression level in
a biological
sample from a subject and comparing the value with the determined standard
value for the
control.
[000299] Expression levels of marker genes include transcription of the marker
genes to
mRNA, and translation into proteins. Therefore, one method of testing for a
disorder and/or
disease is performed based on a comparison of the intensity of expression of
mRNA
corresponding to the marker genes, or the expression level of proteins encoded
by the marker
genes.
[000300] The measurement of the expression levels of marker genes in the
testing for a
disorder and/or disease can be carried out according to various gene analysis
methods.
Specifically, one can use, for example, a hybridization technique using
nucleic acids that
hybridize to these genes as probes, or a gene amplification technique using
DNA that
hybridize to the marker genes as primers.
[000301] The probes or primers used for the testing can be designed based on
the nucleotide
sequences of the marker genes. The identification numbers for the nucleotide
sequences of
the respective marker genes are describer herein.
[000302] Further, it is to be understood that genes of higher animals
generally accompany
polymorphism in a high frequency. There are also many molecules that produce
isoforms
comprising mutually different amino acid sequences during the splicing
process. Any gene
associated with a colon cancer-related disease that has an activity similar to
that of a marker
gene is included in the marker genes, even if it has nucleotide sequence
differences due to
polymorphism or being an isoform.
[000303] It is also to be understood that the marker genes can include
homologs of other
species in addition to humans. Thus, unless otherwise specified, the
expression "marker gene"
refers to a homolog of the marker gene unique to the species or a foreign
marker gene which
has been introduced into an individual.
[000304] Also, it is to be understood that a "homolog of a marker gene" refers
to a gene
derived from a species other than a human, which can hybridize to the human
marker gene as
a probe under stringent conditions. Such stringent conditions are known to one
skilled in the
art who can select an appropriate condition to produce an equal stringency
experimentally or
empirically.

51


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000305] A polynucleotide comprising the nucleotide sequence of a marker gene
or a
nucleotide sequence that is complementary to the complementary strand of the
nucleotide
sequence of a marker gene and has at least 15 nucleotides, can be used as a
primer or probe.
Thus, a "complementary strand" means one strand of a double stranded DNA with
respect to
the other strand and which is composed of A:T (U for RNA) and G:C base pairs.
[000306] In addition, "complementary" means not only those that are completely
complementary to a region of at least 15 continuous nucleotides, but also
those that have a
nucleotide sequence homology of at least 40% in certain instances, 50% in
certain instances,
60% in certain instances, 70% in certain instances, at least 80%, 90%, and 95%
or higher.
The degree of homology between nucleotide sequences can be determined by an
algorithm,
BLAST, etc.
[000307] Such polynucleotides are useful as a probe to detect a marker gene,
or as a primer
to amplify a marker gene. When used as a primer, the polynucleotide comprises
usually 15 bp
to 100 bp, and in certain embodiments 15 bp to 35 bp of nucleotides. When used
as a probe, a
DNA comprises the whole nucleotide sequence of the marker gene (or the
complementary
strand thereof), or a partial sequence thereof that has at least 15 bp
nucleotides. When used as
a primer, the 3' region must be complementary to the marker gene, while the 5'
region can be
linked to a restriction enzyme-recognition sequence or a tag.
[000308] "Polynucleotides" may be either DNA or RNA. These polynucleotides may
be
either synthetic or naturally-occurring. Also, DNA used as a probe for
hybridization is
usually labeled. Those skilled in the art readily understand such labeling
methods. Herein,
the term "oligonucleotide" means a polynucleotide with a relatively low degree
of
polymerization. Oligonucleotides are included in polynucleotides.
[000309] Tests for a disorder and/or disease using hybridization techniques
can be
performed using, for example, Northern hybridization, dot blot hybridization,
or the DNA
microarray technique. Furthermore, gene amplification techniques, such as the
RT-PCR
method may be used. By using the PCR amplification monitoring method during
the gene
amplification step in RT-PCR, one can achieve a more quantitative analysis of
the expression
of a marker gene.
[000310] In the PCR gene amplification monitoring method, the detection target
(DNA or
reverse transcript of RNA) is hybridized to probes that are labeled with a
fluorescent dye and
a quencher which absorbs the fluorescence. When the PCR proceeds and Taq
polymerase
degrades the probe with its 5'-3' exonuclease activity, the fluorescent dye
and the quencher

52


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
draw away from each other and the fluorescence is detected. The fluorescence
is detected in
real time. By simultaneously measuring a standard sample in which the copy
number of a
target is known, it is possible to determine the copy number of the target in
the subject sample
with the cycle number where PCR amplification is linear. Also, one skilled in
the art
recognizes that the PCR amplification monitoring method can be carried out
using any
suitable method.
[000311] The method of testing for a colon cancer-related disease can be also
carried out by
detecting a protein encoded by a marker gene. Hereinafter, a protein encoded
by a marker
gene is described as a "marker protein." For such test methods, for example,
the Western
blotting method, the immunoprecipitation method, and the ELISA method may be
employed
using an antibody that binds to each marker protein.
[000312] Antibodies used in the detection that bind to the marker protein may
be produced
by any suitable technique. Also, in order to detect a marker protein, such an
antibody may be
appropriately labeled. Alternatively, instead of labeling the antibody, a
substance that
specifically binds to the antibody, for example, protein A or protein G, may
be labeled to
detect the marker protein indirectly. More specifically, such a detection
method can include
the ELISA method.
[000313] A protein or a partial peptide thereof used as an antigen may be
obtained, for
example, by inserting a marker gene or a portion thereof into an expression
vector,
introducing the construct into an appropriate host cell to produce a
transformant, culturing the
transformant to express the recombinant protein, and purifying the expressed
recombinant
protein from the culture or the culture supernatant. Alternatively, the amino
acid sequence
encoded by a gene or an oligopeptide comprising a portion of the amino acid
sequence
encoded by a full-length cDNA are chemically synthesized to be used as an
immunogen.
[000314] Furthermore, a test for a colon cancer-related disease can be
performed using as an
index not only the expression level of a marker gene but also the activity of
a marker protein
in a biological sample. Activity of a marker protein means the biological
activity intrinsic to
the protein. Various methods can be used for measuring the activity of each
protein.
[000315] Even if a subject is not diagnosed as being affected with a disorder
and/or disease
in a routine test in spite of symptoms suggesting these diseases, whether or
not such a subject
is suffering from a disorder and/or disease can be easily determined by
performing a test
according to the methods described herein.

53


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000316] More specifically, in certain embodiments, when the marker gene is
one of the
genes described herein, an increase or decrease in the expression level of the
marker gene in a
subject whose symptoms suggest at least a susceptibility to a disorder and/or
disease indicates
that the symptoms are primarily caused thereby.
[000317] In addition, the tests are useful to determine whether a disorder
and/or disease is
improving in a subject. In other words, the methods described herein can be
used to judge the
therapeutic effect of a treatment therefor. Furthermore, when the marker gene
is one of the
genes described herein, an increase or decrease in the expression level of the
marker gene in a
subject, who has been diagnosed as being affected thereby, implies that the
disease has
progressed more.
[000318] The severity and/or susceptibility to a disorder and/or disease may
also be
determined based on the difference in expression levels. For example, when the
marker gene
is one of the genes described herein, the degree of increase in the expression
level of the
marker gene is correlated with the presence and/or severity of a disorder
and/or disease.
[000319] Animal Models
[000320] Animal models for a disorder and/or disease where the expression
level of one or
more marker genes or a gene functionally equivalent to the marker gene has
been elevated in
the animal model can also be made. A "functionally equivalent gene" as used
herein generally
is a gene that encodes a protein having an activity similar to a known
activity of a protein
encoded by the marker gene. A representative example of a functionally
equivalent gene
includes a counterpart of a marker gene of a subject animal, which is
intrinsic to the animal.
[000321] The animal model is useful for detecting physiological changes due to
a disorder
and/or disease. In certain embodiments, the animal model is useful to reveal
additional
functions of marker genes and to evaluate drugs whose targets are the marker
genes.
[000322] An animal model can be created by controlling the expression level of
a
counterpart gene or administering a counterpart gene. The method can include
creating an
animal model by controlling the expression level of a gene selected from the
group of genes
described herein. In another embodiment, the method can include creating an
animal model
by administering the protein encoded by a gene described herein, or
administering an antibody
against the protein. It is to be also understood, that in certain other
embodiments, the marker
can be over-expressed such that the marker can then be measured using
appropriate methods.
In another embodiment, an animal model can be created by introducing a gene
selected from
such groups of genes, or by administering a protein encoded by such a gene. In
another

54


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
embodiment, a disorder and/or disease can be induced by suppressing the
expression of a gene
selected from such groups of genes or the activity of a protein encoded by
such a gene. An
antisense nucleic acid, a ribozyme, or an RNAi can be used to suppress the
expression. The
activity of a protein can be controlled effectively by administering a
substance that inhibits the
activity, such as an antibody.
[000323] The animal model is useful to elucidate the mechanism underlying a
disorder
and/or disease and also to test the safety of compounds obtained by screening.
For example,
when an animal model develops the symptoms of a particular disorder and/or
disease, or when
a measured value involved in a certain a disorder and/or disease alters in the
animal, a
screening system can be constructed to explore compounds having activity to
alleviate the
disease.
[000324] As used herein, the expression "an increase in the expression level"
refers to any
one of the following: where a marker gene introduced as a foreign gene is
expressed
artificially; where the transcription of a marker gene intrinsic to the
subject animal and the
translation thereof into the protein are enhanced; or where the hydrolysis of
the protein, which
is the translation product, is suppressed.
[000325] As used herein, the expression "a decrease in the expression level"
refers to either
the state in which the transcription of a marker gene of the subject animal
and the translation
thereof into the protein are inhibited, or the state in which the hydrolysis
of the protein, which
is the translation product, is enhanced. The expression level of a gene can be
determined, for
example, by a difference in signal intensity on a DNA chip. Furthermore, the
activity of the
translation product--the protein--can be determined by comparing with that in
the normal
state.
[000326] It is also within the contemplated scope that the animal model can
include
transgenic animals, including, for example animals where a marker gene has
been introduced
and expressed artificially; marker gene knockout animals; and knock-in animals
in which
another gene has been substituted for a marker gene. A transgenic animal, into
which an
antisense nucleic acid of a marker gene, a ribozyme, a polynucleotide having
an RNAi effect,
or a DNA functioning as a decoy nucleic acid or such has been introduced, can
be used as the
transgenic animal. Such transgenic animals also include, for example, animals
in which the
activity of a marker protein has been enhanced or suppressed by introducing a
mutation(s) into
the coding region of the gene, or the amino acid sequence has been modified to
become



CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
resistant or susceptible to hydrolysis. Mutations in an amino acid sequence
include
substitutions, deletions, insertions, and additions.
[000327] Examples of Expression
[000328] In addition, the expression itself of a marker gene can be controlled
by introducing
a mutation(s) into the transcriptional regulatory region of the gene. Those
skilled in the art
understand such amino acid substitutions. Also, the number of amino acids that
are mutated is
not particularly restricted, as long as the activity is maintained. Normally,
it is within 50
amino acids, in certain non-limiting embodiments, within 30 amino acids,
within 10 amino
acids, or within 3 amino acids. The site of mutation may be any site, as long
as the activity is
maintained.
[000329] In yet another aspect, there is provided herein screening methods for
candidate
compounds for therapeutic agents to treat a particular disorder and/or
disease. One or more
marker genes are selected from the group of genes described herein. A
therapeutic agent for a
colon cancer-related disease can be obtained by selecting a compound capable
of increasing or
decreasing the expression level of the marker gene(s).
[000330] It is to be understood that the expression "a compound that increases
the
expression level of a gene" refers to a compound that promotes any one of the
steps of gene
transcription, gene translation, or expression of a protein activity. On the
other hand, the
expression "a compound that decreases the expression level of a gene", as used
herein, refers
to a compound that inhibits any one of these steps.
[000331] In particular aspects, the method of screening for a therapeutic
agent for a disorder
and/or disease can be carried out either in vivo or in vitro. This screening
method can be
performed, for example, by:

1) administering a candidate compound to an animal subject;

2) measuring the expression level of a marker gene(s) in a biological sample
from
the animal subject; or

3) selecting a compound that increases or decreases the expression level of a
marker gene(s) as compared to that in a control with which the candidate
compound has
not been contacted.
[000332] In still another aspect, there is provided herein a method to assess
the efficacy of a
candidate compound for a pharmaceutical agent on the expression level of a
marker gene(s)
by contacting an animal subject with the candidate compound and monitoring the
effect of the

56


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
compound on the expression level of the marker gene(s) in a biological sample
derived from
the animal subject. The variation in the expression level of the marker
gene(s) in a biological
sample derived from the animal subject can be monitored using the same
technique as used in
the testing method described above. Furthermore, based on the evaluation, a
candidate
compound for a pharmaceutical agent can be selected by screening.
[000333] All patents, patent applications and references cited herein are
incorporated in their
entirety by reference. While the invention has been described and exemplified
in sufficient
detail for those skilled in this art to make and use it, various alternatives,
modifications and
improvements should be apparent without departing from the spirit and scope of
the invention.
One skilled in the art readily appreciates that the present invention is well
adapted to carry out
the objects and obtain the ends and advantages mentioned, as well as those
inherent therein.
[000334] Certain Nucleobase Sequences
[000335] Nucleobase sequences of mature miRNAs and their corresponding stem-
loop
sequences described herein are the sequences found in miRBase, an online
searchable
database of miRNA sequences and annotation, found at
http://microma.sanger.ac.uk/. Entries
in the miRBase Sequence database represent a predicted hairpin portion of a
miRNA
transcript (the stem-loop), with information on the location and sequence of
the mature
miRNA sequence. The miRNA stem-loop sequences in the database are not strictly
precursor
miRNAs (pre-miRNAs), and may in some instances include the pre-miRNA and some
flanking sequence from the presumed primary transcript. The miRNA nucleobase
sequences
described herein encompass any version of the miRNA, including the sequences
described in
Release 10.0 of the miRBase sequence database and sequences described in any
earlier
Release of the miRBase sequence database. A sequence database release may
result in the re-
naming of certain miRNAs. A sequence database release may result in a
variation of a mature
miRNA sequence. The compounds that may encompass such modified
oligonucleotides may
be complementary any nucleobase sequence version of the miRNAs described
herein.
[000336] It is understood that any nucleobase sequence set forth herein is
independent of any
modification to a sugar moiety, an internucleoside linkage, or a nucleobase.
It is further
understood that a nucleobase sequence comprising U's also encompasses the same
nucleobase
sequence wherein 'U' is replaced by 'T' at one or more positions having 'U."
Conversely, it
is understood that a nucleobase sequence comprising T's also encompasses the
same
nucleobase sequence wherein 'T; is replaced by 'U' at one or more positions
having 'T."

57


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000337] In certain embodiments, a modified oligonucleotide has a nucleobase
sequence that
is complementary to a miRNA or a precursor thereof, meaning that the
nucleobase sequence
of a modified oligonucleotide is a least 60%, 65%, 70%, 75%, 80%, 85%, 90%,
95%, 97%,
98% or 99% identical to the complement of a miRNA or precursor thereof over a
region of 8,
9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40,
45, 50, 55, 60, 65,
70, 75, 80, 85, 90, 95, 100 or more nucleobases, or that the two sequences
hybridize under
stringent hybridization conditions. Accordingly, in certain embodiments the
nucleobase
sequence of a modified oligonucleotide may have one or more mismatched
basepairs with
respect to its target miRNA or target miRNA precursor sequence, and is capable
of
hybridizing to its target sequence. In certain embodiments, a modified
oligonucleotide has a
nucleobase sequence that is 100% complementary to a miRNA or a precursor
thereof. In
certain embodiments, the nucleobase sequence of a modified oligonucleotide has
full-length
complementary to a miRNA.
[000338] miRNA (miR) Therapies
[000339] In some embodiments, the present invention provides microRNAs that
inhibit the
expression of one or more genes in a subject. MicroRNA expression profiles can
serve as a
new class of cancer biomarkers.
[000340] Included herein are methods of inhibiting gene expression and/or
activity using one
or more MiRs. In some embodiments, the miR(s) inhibit the expression of a
protein. In other
embodiments, the miRNA(s) inhibits gene activity (e.g., cell invasion
activity).
[000341] The miRNA can be isolated from cells or tissues, recombinantly
produced, or
synthesized in vitro by a variety of techniques well known to one of ordinary
skill in the art.
In one embodiment, miRNA is isolated from cells or tissues. Techniques for
isolating miRNA
from cells or tissues are well known to one of ordinary skill in the art. For
example, miRNA
can be isolated from total RNA using the mirVana miRNA isolation kit from
Ambion, Inc.
Another techniques utilizes the flashIPAGETM Fractionator System (Ambion,
Inc.) for PAGE
purification of small nucleic acids.
[000342] For the use of miRNA therapeutics, it is understood by one of
ordinary skill in the
art that nucleic acids administered in vivo are taken up and distributed to
cells and tissues.
[000343] The nucleic acid may be delivered in a suitable manner which enables
tissue-
specific uptake of the agent and/or nucleic acid delivery system. The
formulations described
herein can supplement treatment conditions by any known conventional therapy,
including,
but not limited to, antibody administration, vaccine administration,
administration of cytotoxic

58


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
agents, natural amino acid polypeptides, nucleic acids, nucleotide analogues,
and biologic
response modifiers. Two or more combined compounds may be used together or
sequentially.
[000344] Certain embodiments of the invention provide pharmaceutical
compositions
containing (a) one or more nucleic acid or small molecule compounds and (b)
one or more
other chemotherapeutic agents.
[000345] Additional Useful Definitions
[000346] "Subject" means a human or non-human animal selected for treatment or
therapy.
Subject suspected of having" means a subject exhibiting one or more clinical
indicators of a
disorder, disease or condition.
[000347] Preventing" or "prevention" refers to delaying or forestalling the
onset,
development or progression of a condition or disease for a period of time,
including weeks,
months, or years. Treatment" or "treat" means the application of one or more
specific
procedures used for the cure or amelioration of a disorder and/or disease. In
certain
embodiments, the specific procedure is the administration of one or more
pharmaceutical
agents.
[000348] "Amelioration" means a lessening of severity of at least one
indicator of a
condition or disease. In certain embodiments, amelioration includes a delay or
slowing in the
progression of one or more indicators of a condition or disease. The severity
of indicators
may be determined by subjective or objective measures which are known to those
skilled in
the art.
[000349] Subject in need thereof" means a subject identified as in need of a
therapy or
treatment.
[000350] "Administering" means providing a pharmaceutical agent or composition
to a
subject, and includes, but is not limited to, administering by a medical
professional and self-
administering.
[000351] "Parenteral administration," means administration through injection
or infusion.
Parenteral administration includes, but is not limited to, subcutaneous
administration,
intravenous administration, intramuscular administration, intraarterial
administration, and
intracranial administration. Subcutaneous administration" means administration
just below
the skin.
[000352] "Improves function" means the changes function toward normal
parameters. In
certain embodiments, function is assessed by measuring molecules found in a
subject's bodily
fluids. Pharmaceutical composition" means a mixture of substances suitable for
administering

59


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
to an individual that includes a pharmaceutical agent. For example, a
pharmaceutical
composition may comprise a modified oligonucleotide and a sterile aqueous
solution.
[000353] "Target nucleic acid," "target RNA," "target RNA transcript" and
"nucleic acid
target" all mean a nucleic acid capable of being targeted by antisense
compounds. Targeting"
means the process of design and selection of nucleobase sequence that will
hybridize to a
target nucleic acid and induce a desired effect. "Targeted to" means having a
nucleobase
sequence that will allow hybridization to a target nucleic acid to induce a
desired effect. In
certain embodiments, a desired effect is reduction of a target nucleic acid.
[000354] "Modulation" means to a perturbation of function or activity. In
certain
embodiments, modulation means an increase in gene expression. In certain
embodiments,
modulation means a decrease in gene expression.
[000355] "Expression" means any functions and steps by which a gene's coded
information
is converted into structures present and operating in a cell.
[000356] "Region" means a portion of linked nucleosides within a nucleic acid.
In certain
embodiments, a modified oligonucleotide has a nucleobase sequence that is
complementary to
a region of a target nucleic acid. For example, in certain such embodiments a
modified
oligonucleotide is complementary to a region of a miRNA stem-loop sequence. In
certain
such embodiments, a modified oligonucleotide is 100% identical to a region of
a miRNA
sequence.
[000357] "Segment" means a smaller or sub-portion of a region.
[000358] "Nucleobase sequence" means the order of contiguous nucleobases, in a
5' to 3'
orientation, independent of any sugar, linkage, and/or nucleobase
modification.
[000359] Contiguous nucleobases" means nucleobases immediately adjacent to
each other in
a nucleic acid.
[000360] "Nucleobase complementarity" means the ability of two nucleobases to
pair non-
covalently via hydrogen bonding. "Complementary" means a first nucleobase
sequence is at
least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% identical, or is
100%
identical, to the complement of a second nucleobase sequence over a region of
8, 9, 10, 11, 12,
13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 55,
60, 65, 70, 75, 80, 85,
90, 95, 100 or more nucleobases, or that the two sequences hybridize under
stringent
hybridization conditions. In certain embodiments a modified oligonucleotide
that has a
nucleobase sequence which is 100% complementary to a miRNA, or precursor
thereof, may



CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
not be 100% complementary to the miRNA, or precursor thereof, over the entire
length of the
modified oligonucleotide.
[000361] "Complementarity" means the nucleobase pairing ability between a
first nucleic
acid and a second nucleic acid. "Full-length complementarity" means each
nucleobase of a
first nucleic acid is capable of pairing with each nucleobase at a
corresponding position in a
second nucleic acid. For example, in certain embodiments, a modified
oligonucleotide
wherein each nucleobase has complementarity to a nucleobase in an miRNA has
full-length
complementarity to the miRNA.
[000362] "Percent complementary" means the number of complementary nucleobases
in a
nucleic acid divided by the length of the nucleic acid. In certain
embodiments, percent
complementarity of a modified oligonucleotide means the number of nucleobases
that are
complementary to the target nucleic acid, divided by the number of nucleobases
of the
modified oligonucleotide. In certain embodiments, percent complementarity of a
modified
oligonucleotide means the number of nucleobases that are complementary to a
miRNA,
divided by the number of nucleobases of the modified oligonucleotide.
[000363] "Percent region bound" means the percent of a region complementary to
an
oligonucleotide region. Percent region bound is calculated by dividing the
number of
nucleobases of the target region that are complementary to the oligonucleotide
by the length
of the target region. In certain embodiments, percent region bound is at least
80%, at least
85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at
least 99%, or
100%.
[000364] "Percent identity" means the number of nucleobases in first nucleic
acid that are
identical to nucleobases at corresponding positions in a second nucleic acid,
divided by the
total number of nucleobases in the first nucleic acid.
[000365] "Substantially identical" used herein may mean that a first and
second nucleobase
sequence are at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99%
identical, or 100% identical, over a region of 8, 9, 10, 11, 12, 13, 14, 15,
16, 17, 18, 19, 20, 21,
22, 23, 24, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or
more nucleobases.
[000366] "Hybridize" means the annealing of complementary nucleic acids that
occurs
through nucleobase complementarity.
[000367] "Mismatch" means a nucleobase of a first nucleic acid that is not
capable of pairing
with a nucleobase at a corresponding position of a second nucleic acid.

61


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000368] "Non-complementary nucleobase" means two nucleobases that are not
capable of
pairing through hydrogen bonding.
[000369] "Identical" means having the same nucleobase sequence.
[000370] "miRNA" or "miR" means a non-coding RNA between 18 and 25 nucleobases
in
length which hybridizes to and regulates the expression of a coding RNA. In
certain
embodiments, a miRNA is the product of cleavage of a pre-miRNA by the enzyme
Dicer.
Examples of miRNAs are found in the miRNA database known as miRBase
(http://microrna. sanger. ac.uk/).
[000371] "Pre-miRNA" or "pre-miR" means a non-coding RNA having a hairpin
structure,
which contains a miRNA. In certain embodiments, a pre-miRNA is the product of
cleavage of
a pri-miR by the double-stranded RNA-specific ribonuclease known as Drosha.
[000372] "Stem-loop sequence" means an RNA having a hairpin structure and
containing a
mature miRNA sequence. Pre-miRNA sequences and stem-loop sequences may
overlap.
Examples of stem-loop sequences are found in the miRNA database known as
miRBase
(http://microrna. sanger. ac.uk/.
[000373] "miRNA precursor" means a transcript that originates from a genomic
DNA and
that comprises a non-coding, structured RNA comprising one or more miRNA
sequences.
For example, in certain embodiments a miRNA precursor is a pre-miRNA. In
certain
embodiments, a miRNA precursor is a pri-miRNA.
[000374] "Antisense compound" means a compound having a nucleobase sequence
that will
allow hybridization to a target nucleic acid. In certain embodiments, an
antisense compound
is an oligonucleotide having a nucleobase sequence complementary to a target
nucleic acid.
[000375] "Oligonucleotide" means a polymer of linked nucleosides, each of
which can be
modified or unmodified, independent from one another. "Naturally occurring
internucleoside
linkage" means a 3' to 5' phosphodiester linkage between nucleosides. "Natural
nucleobase"
means a nucleobase that is unmodified relative to its naturally occurring
form. "miR
antagonist" means an agent designed to interfere with or inhibit the activity
of a miRNA. In
certain embodiments, a miR antagonist comprises an antisense compound targeted
to a
miRNA. In certain embodiments, a miR antagonist comprises a modified
oligonucleotide
having a nucleobase sequence that is complementary to the nucleobase sequence
of a miRNA,
or a precursor thereof. In certain embodiments, an miR antagonist comprises a
small
molecule, or the like that interferes with or inhibits the activity of an
miRNA.

62


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
[000376] The methods and reagents described herein are representative of
preferred
embodiments, are exemplary, and are not intended as limitations on the scope
of the invention.
Modifications therein and other uses will occur to those skilled in the art.
These modifications
are encompassed within the spirit of the invention and are defined by the
scope of the claims.
It will also be readily apparent to a person skilled in the art that varying
substitutions and
modifications may be made to the invention disclosed herein without departing
from the scope
and spirit of the invention.
[000377] It should be understood that although the present invention has been
specifically
disclosed by preferred embodiments and optional features, modifications and
variations of the
concepts herein disclosed may be resorted to by those skilled in the art, and
that such
modifications and variations are considered to be within the scope of this
invention as defined
by the appended claims.
[000378] While the invention has been described with reference to various and
preferred
embodiments, it should be understood by those skilled in the art that various
changes may be
made and equivalents may be substituted for elements thereof without departing
from the
essential scope of the invention. In addition, many modifications may be made
to adapt a
particular situation or material to the teachings of the invention without
departing from the
essential scope thereof.
[000379] REFERENCES
[000380] The publication and other material used herein to illuminate the
invention or
provide additional details respecting the practice of the invention, are
incorporated by
reference herein, and for convenience are provided in the following
bibliography.
[000381] Citation of the any of the documents recited herein is not intended
as an admission
that any of the foregoing is pertinent prior art. All statements as to the
date or representation
as to the contents of these documents is based on the information available to
the applicant
and does not constitute any admission as to the correctness of the dates or
contents of these
documents.
[000382]
Arata, Y., Fujita, M., Ohtani, K., Kijima, S., and Kato, J. Y. (2000). Cdk2-
dependent and
- independent pathways in E2F-mediated S phase induction. J Biol Chem 275,
6337-
6345. Attwooll, C., Lazzerini Denchi, E., and Helin, K. (2004). The E2F
family:
specific functions and overlapping interests. EMBO J 23, 4709-4716.
Blow, J. J., and Hodgson, B. (2002). Replication licensing--defining the
proliferative
63


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
state? Trends Cell Biol 12, 72-78.
Bonci, D., Cittadini, A., Latronico, M. V. G., Borello, U., Aycock, J. K.,
Drusco, A.,
Innocenzi, A., Follenzi, A., Lavitrano, M., Monti, M. G., et al. (2003).
'Advanced'
generation lentiviruses as efficient vectors for cardiomyocyte gene
transduction in
vitro and in vivo. Gene Therapy 10, 630636.
Costinean, S., Zanesi, N., Pekarsky, Y., Tili, E., Volinia, S., Heerema, N.,
and Croce, C.
M. (2006). Pre-B cell proliferation and lymphoblastic leukemia/high-grade
lymphoma
in E(mu)miR155 transgenic mice. Proc Natl Acad Sci U S A 103, 7024-7029.
De Caestecker, M. P., Piek, E., and Roberts, A. B. (2000). Role of
transforming growth
factor-beta signaling in cancer. J Natl Cancer Inst 92, 1388-1402.
DeGregori, J. (2002). The genetics of the E2F family of transcription factors:
shared
functions and unique roles. Biochim Biophys Acta 1602, 131-150.

Dews, M., Homayouni, A., Yu, D., Murphy, D., Sevignani, C., Wentzel, E.,
Furth, E. E.,
Lee, W. M., Enders, G. H., Mendell, J. T., and Thomas-Tikhonenko, A. (2006).
Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster. Nat
Genet 38, 1060-1065.
Dyson, N. (1998). The regulation of E2F by pRB-family proteins. Genes Dev 12,
2245-
2262.
Egle, A., Harris, A. W., Bouillet, P., and Cory, S. (2004). Bim is a
suppressor of Myc-
induced mouse B cell leukemia. Proc Natl Acad Sci 101, 6164-6169.
Esquela-Kerscher A, Slack FJ. (2006) Oncomirs - microRNAs with a role in
cancer. Nat
Rev Cancer. 6, 259-69
Fox, J. G., Beck, P., Dangler, C. A., Whary, M. T., Wang, T. C., Shi, H. N.,
and Nagler-
Anderson, C. (2000). Concurrent enteric helminth infection modulates
inflammation
and gastric immune responses and reduces helicobacter-induced gastric atrophy.
Nat
Med 6, 536-542. Gross, A., McDonnell, J. M., and Korsmeyer, S. J. (1999). BCL-
2
family members and the mitochondria in apoptosis. Genes Dev 13, 1899-1911.

He, L., Thomson, J. M., Hemann, M. T., Hernando-Monge, E., Mu, D., Goodson,
S.,
Powers, S., Cordon-Cardo, C., Lowe, S. W., Hannon, G. J., and Hammond, S. M.
(2005). A microRNA polycistron as a potential human oncogene. Nature 435, 828-
833.
Hossain, A., Kuo, M. T., and Saunders, G. F. (2006). Mir-17-5p regulates
breast cancer
cell proliferation by inhibiting translation of AIB 1 mRNA. Mol Cell Biol 26,
8191-
64


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
8201.

Houbaviy, H. B., Murray, M. F., and Sharp, P. A. (2003). Embryonic stem cell-
specific
MicroRNAs. Dev Cell 5, 351-358.
Houghton, J., Fox, J. G., and Wang, T. C. (2002). Gastric cancer: laboratory
bench to
clinic. J Gastroenterol Hepatol 17, 495-502.
Hwang, H. W., Wentzel, E. A., and Mendell, J. T. (2007). A hexanucleotide
element
directs microRNA nuclear import. Science 315, 97-100.
Johnson, D. G., and DeGregori, J. (2006). Putting the Oncogenic and Tumor
Suppressive
Activities of E2F into Context. Curr Mol Med 6, 731-738.

Ju, H. R., Jung, U., Sonn, C. H., Yoon, S. R., Jeon, J. H., Yang, Y., Lee, K.
N., and Choi,
1. (2003). Aberrant signaling of TGF-betal by the mutant Smad4 in gastric
cancer
cells. Cancer Lett 196, 197-206.

Kim, Y.K. and Kim, V.K. (2007). Processing of intronic microRNAs. EMBO J 26,
775-
783.
Kloosterman, W. P., and Plasterk, R. H. (2006). The diverse functions of
microRNAs in
animal development and disease. Dev Cell 11, 441-450.
Lauren P (1965) The two histological main types of gastric carcinoma: Diffuse
and so-
called intestinal-type carcinoma. An attempt at a histo-clnical
classification. Acta
Pathol Microbiol Scand 64: 31-49.
Lazzerini Denchi, E., and Helin, K. (2005). E2F1 is crucial for E2F-dependent
apoptosis.
EMBO Rep 6, 661-668.
Leone, G., DeGregori, J., Yan, Z., Jakoi, L., Ishida, S., Williams, R. S., and
Nevins, J. R.
(1998). E2F3 activity is regulated during the cell cycle and is required for
the
induction of S phase. Genes Dev 12, 2120-2130.

Lewis, B. P., Burge, C. B., and Bartel, D. P. (2005). Conserved Seed Pairing,
Often
Flanked by Adenosines, Indicates that Thousands of Human Genes are MicroRNA
Targets. Cell 120 15-20. Li, S. S., Xue, W. C., Khoo, U. S., Ngan, H. Y.,
Chan, K. Y.,
Tam, I. Y., Chiu, P. M., Ip, P. P., Tam, K. F., and Cheung, A. N. (2005).
Replicative
MCM7 protein as a proliferation marker in endometrial carcinoma: a tissue
microarray
and clinicopathological analysis. Histopathology 46, 307-313.
Liu, C.-G., Calin, G. A., Meloon, B., Gamliel, N., Sevignani, C., Ferracin,
M., Dumitru, C.
D., Shimizu, M., Zupo, S., Dono, M., et al. (2004). An oligonucleotide
microchip for
genome-wide microRNA profiling in human and mouse tissues. Proc Natl Acad Sci



CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
101, 9740-9744. Lu, J., Getz, G., Miska, E. A., Alvarez-Saavedra, E., Lamb,
J., Peck,
D., Sweet-Cordero, A., Ebert, B. L., Mak, R. H., Ferrando, A. A., et al.
(2005).
MicroRNA expression profiles classify human cancers. Nature 435, 834-838.
Ma L., Teruya-Feldstein J. and Weinberg R.A. (2007) Tumour invasion and
metastasis
initiated by microRNA- I Ob in breast cancer. Nature. 449, 682-8.
Mattioli, E., Vogiatzi, P., Sun, A., Abbadessa, G., Angeloni, G., D'Ugo, D.,
Trani, D.,
Gaughan, J. P., Vecchio, F. M., Cevenini, G., et al. (2007).
Immunohistochemical
analysis of pRb2/p130, VEGF, EZH2, p53, p16(INK4A), p27(KIP1), p21(WAF1), Ki-
67 expression patterns in gastric cancer. J Cell Physiol 210, 183-191.
Meng, F., Henson, R., Lang, M., Wehbe, H., Maheshwari, S., Mendell, J. T.,
Jiang, J.,
Schmittgen, T. D., and Patel, T. (2006). Involvement of human micro-RNA in
growth
and response to chemotherapy in human cholangiocarcinoma cell lines.
Gastroenterology 130, 21132129.
Nahle, Z., Polakoff, J., Davuluri, R. V., McCurrach, M. E., Jacobson, M. D.,
Narita, M.,
Zhang, M. Q., Lazebnik, Y., Bar-Sagi, D., and Lowe, S. W. (2002). Direct
coupling of
the cell cycle and cell death machinery by E2F. Nat Cell Biol 4, 859-864.
O'Donnell, K. A., Wentzel, E. A., Zeller, K. I., Dang, C. V., and Mendell, J.
T. (2005). c-
Myc-regulated microRNAs modulate E2F1 expression. Nature 435, 839-843.
Ohgushi, M., Kuroki, S., Fukamachi, H., O'Reilly, L. A., Kuida, K., Strasser,
A., and
Yonehara, S. (2005). Transforming growth factor beta-dependent sequential
activation
of Smad, Bim, and caspase-9 mediates physiological apoptosis in gastric
epithelial
cells. Mol Cell Biol 25, 1001710028.
Park, K., Kim, S. J., Bang, Y. J., Park, J. G., Kim, N. K., Roberts, A. B.,
and Sporn, M. B.
(1994). Genetic changes in the transforming growth factor beta (TGF-beta) type
II
receptor gene in human gastric cancer cells: correlation with sensitivity to
growth
inhibition by TGF-beta. Proc Natl Acad Sci U S A 91, 8772-8776.
Peng, D. F., Sugihara, H., Mukaisho, K., Tsubosa, Y., and Hattori, T. (2003).
Alterations
of chromosomal copy number during progression of diffuse-type gastric
carcinomas:
metaphase- and array-based comparative genomic hybridization analyses of
multiple
samples from individual tumours. J Pathol 201, 439-450.

Pierce, A. M., Schneider-Broussard, R., Gimenez-Conti, I. B., Russell, J. L.,
Conti, C. J.,
and Johnson, D. G. (1999). E2F1 has both oncogenic and tumor-suppressive
properties
in a transgenic model. Mol Cell Biol 19, 6408-6414.

66


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
Ren, B., Yu, G., Tseng, G. C., Cieply, K., Gavel, T., Nelson, J.,
Michalopoulos, G., Yu, Y.
P., and Luo, J. H. (2006). MCM7 amplification and overexpression are
associated with
prostate cancer progression. Oncogene 25, 1090-1098.
Rodriguez, A., Vigorito, E., Clare, S., Warren, M. V., Couttet, P., Soond, D.
R., van
Dongen, S., Grocock, R. J., Das, P. P., Miska, E. A., et al. (2007).
Requirement of
bic/microRNA- 155 for normal immune function. Science 316, 608-611.
Scandura, J. M., Boccuni, P., Massague, J., and Nimer, S. D. (2004).
Transforming growth
factor beta-induced cell cycle arrest of human hematopoietic cells requires
p57KIP2
up-regulation. Proc Natl Acad Sci U S A 101, 15231-15236.
Suzuki, S., Adachi, A., Hiraiwa, A., Ohashi, M., Ishibashi, M., and Kiyono, T.
(1998).
Cloning and characterization of human MCM7 promoter. Gene 216, 85-91.
Suzuki, T., Yasui, W., Yokozaki, H., Naka, K., Ishikawa, T., and Tahara, E.
(1999).
Expression of the E2F family in human gastrointestinal carcinomas. Int J
Cancer 81,
535-538.
Sylvestre, Y., De Guire, V., Querido, E., Mukhopadhyay, U. K., Bourdeau, V.,
Major, F.,
Ferbeyre, G., and Chartrand, P. (2007). An E2F/miR-20a autoregulatory feedback
loop. J Biol Chem 282, 2135-2143.
Takada, H., Imoto, I., Tsuda, H., Sonoda, I., Ichikura, T., Mochizuki, H.,
Okanoue, T., and
Inazawa, J. (2005). Screening of DNA copy-number aberrations in gastric cancer
cell
lines by array-based comparative genomic hybridization. Cancer Sci 96, 100-
110.
Tan, T. T., Degenhardt, K., Nelson, D. A., Beaudoin, B., Nieves-Neira, W.,
Bouillet, P.,
Villunger, A., Adams, J. M., and White, E. (2005). Key roles of BIM-driven
apoptosis
in epithelial tumors and rational chemotherapy. Cancer Cell 7, 227-238.
Thai, T. H., Calado, D. P., Casola, S., Ansel, K. M., Xiao, C., Xue, Y.,
Murphy, A.,
Frendewey, D., Valenzuela, D., Kutok, J. L., et al. (2007). Regulation of the
germinal
center response by microRNA-155. Science 316, 604-608.

Thomson, J. M., Newman, M., Parker, J. S., Morin-Kensicki, E. M., Wright, T.,
and
Hammond, S. M. (2006). Extensive post-transcriptional regulation of microRNAs
and
its implications for cancer. Genes Dev 20, 2202-2207.
Uemura, N., Okamoto, S., Yamamoto, S., Matsumura, N., Yamaguchi, S., Yamakido,
M.,
Taniyama, K., Sasaki, N., and Schlemper, R. J. (2001). Helicobacter pylori
infection
and the development of gastric cancer. N Engl J Med 345, 784-789.
van den Brink, G. R., and Offerhaus, G. J. (2007). The morphogenetic code and
colon
67


CA 02716906 2010-08-26
WO 2009/108853 PCT/US2009/035458
cancer development. Cancer Cell 11, 109-117.
Volinia, S., Calin, G. A., Liu, C.-G., Ambs, S., Cimmino, A., Petrocca, F.,
Visone, R.,
Iorio, M., Roldo, C., Ferracin, M., et al. (2006). A microRNA expression
signature of
human solid tumors defines cancer gene targets. Proc Natl Acad Sci 103, 2257-
2261.
Voorhoeve, P. M., le Sage, C., Schrier, M., Gillis, A. J., Stoop, H., Nagel,
R., Liu, Y. P.,
van Duijse, J., Drost, J., Griekspoor, A., et al. (2006). A genetic screen
implicates
miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell 124,
1169-1181.

Weiss, M. M., Kuipers, E. J., Postma, C., Snijders, A. M., Pinkel, D.,
Meuwissen, S. G.,
Albertson, D., and Meijer, G. A. (2004). Genomic alterations in primary
gastric
adenocarcinomas correlate with clinicopathological characteristics and
survival. Cell
Oncol 26, 307-317.
Woods, K., Thomson, J. M., and Hammond, S. M. (2007). Direct regulation of an
oncogenic micro-RNA cluster by E2F transcription factors. J Biol Chem 282,
2130-
2134.

Yanaihara, N., Caplen, N., Bowman, E., Seike, M., Kumamoto, K., Yi, M.,
Stephens, R.
M., Okamoto, A., Yokota, J., Tanaka, T., et al. (2006). Unique microRNA
molecular
profiles in lung cancer diagnosis and prognosis. Cancer Cell 9, 189-198.
Yano, T., Ito, K., Fukamachi, H., Chi, X. Z., Wee, H. J., Inoue, K., Ida, H.,
Bouillet, P.,
Strasser, A., Bae, S. C., and Ito, Y. (2006). The RUNX3 tumor suppressor
upregulates
Bim in gastric epithelial cells undergoing transforming growth factor beta-
induced
apoptosis. Mol Cell Biol 26, 4474-4488.

68

Representative Drawing

Sorry, the representative drawing for patent document number 2716906 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2009-02-27
(87) PCT Publication Date 2009-09-03
(85) National Entry 2010-08-26
Examination Requested 2014-02-19
Dead Application 2018-12-12

Abandonment History

Abandonment Date Reason Reinstatement Date
2017-12-12 R30(2) - Failure to Respond
2018-02-27 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2010-08-26
Maintenance Fee - Application - New Act 2 2011-02-28 $100.00 2011-02-01
Maintenance Fee - Application - New Act 3 2012-02-27 $100.00 2012-02-24
Maintenance Fee - Application - New Act 4 2013-02-27 $100.00 2013-01-31
Maintenance Fee - Application - New Act 5 2014-02-27 $200.00 2014-01-31
Request for Examination $800.00 2014-02-19
Maintenance Fee - Application - New Act 6 2015-02-27 $200.00 2015-02-05
Maintenance Fee - Application - New Act 7 2016-02-29 $200.00 2016-02-05
Maintenance Fee - Application - New Act 8 2017-02-27 $200.00 2017-01-31
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE OHIO STATE UNIVERSITY RESEARCH FOUNDATION
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2010-08-26 1 52
Claims 2010-08-26 10 379
Drawings 2010-08-26 75 4,383
Description 2010-08-26 68 3,896
Cover Page 2010-12-02 1 29
Description 2015-09-28 68 3,859
Claims 2015-09-28 1 46
Claims 2016-11-23 3 121
Correspondence 2010-10-28 1 29
Examiner Requisition 2017-06-12 6 417
PCT 2010-08-26 10 519
Assignment 2010-08-26 4 129
Correspondence 2010-09-20 1 39
Correspondence 2010-11-26 2 62
Fees 2011-02-01 1 37
Prosecution Correspondence 2014-02-19 3 116
Prosecution-Amendment 2014-02-19 1 37
Prosecution-Amendment 2015-03-26 8 498
Amendment 2015-09-28 27 1,202
Examiner Requisition 2016-05-27 4 301
Amendment 2016-11-23 14 569